MICHELE CORRÊA BERTOLDI
ANTIOXIDANT CAPACITY, ANTICANCER EFFECTS AND ABSORPTION
OF MANGO (Mangifera Indica L.) POLYPHENOLS IN VITRO
Tese apresentada à Universidade
Federal de Viçosa, como parte das
exigências do Programa de PósGraduação em Ciência e Tecnologia de
Alimentos, para obtenção do título de
Doctor Scientiae.
VIÇOSA
MINAS GERAIS-BRASIL
2009
iii
MICHELE CORRÊA BERTOLDI
ANTIOXIDANT CAPACITY, ANTICANCER EFFECTS AND ABSORPTION
OF MANGO (Mangifera Indica L) POLYPHENOLS IN VITRO
Tese apresentada à Universidade
Federal de Viçosa, como parte das
exigências do Programa de PósGraduação em Ciência e Tecnologia de
Alimentos, para obtenção do título de
Doctor Scientiae.
APROVADA: 9 de dezembro de 2009.
_________________________
Profa. Tânia Toledo de Oliveira
(Coorientadora)
_____________________________
Profa.Susanne U Mertens-Talcott
(Coorientadora)
_________________________
Prof. José Carlos Gomes
______________________________
Profa. Nilda de Fátima Ferreira Soares
_____________________________
Prof. Paulo Cesar Stringheta
(Orientador)
To my son Vitor, for his unconditional love.
To my husband Hernani, for his love, care and friendship.
To my family, for their constant support.
ii
ACKNOWLEDGMENTS
I would like to thank GOD, for everything I have, light and protection.
I would like to express my gratitude to my chair Paulo Cesar Stringheta for his
friendship, scientific support, opportunities and assistance during my academic
trajectory.
I would like to thank my co-chairs Susanne U Mertens-Talcott and Stephen T.
Talcott for their scientific support, friendship and the generous assistance with the
Doctoral training in USA. They really help my family and I to overcome the challenge
of living a new culture.
I would like to thank the Universidade Federal de Viçosa and the Department of
Food Science and Technology for the opportunity and also to CAPES Foundation
(Ministry of Education, Brazil) for providing the fellowship to conduct the research
experiment in the USA (Grant n. BEX 130-08-7).
I very appreciate the National Mango Board support for providing plant material
used in this study as well as for the financial support.
I would like to thank the members of my doctoral committee and other
professors for their scientific support. José Carlos Gomes, Nilda de Fátima Ferreira
Soares, Tânia Toledo de Oliveira, Valéria Paula Rodrigues Minim, Afonso Mota
Ramos, Frederico José Vieira Passos, Júlio Maria de Andrade Araújo, Mônica Ribeiro
Pirozzi, José Benício Paes Chaves, among others, have been taught value scientific
background as well as academic skills. I would especially like to thank the professors
Nilda de Fatima Ferreira Soares, Paulo Cesar Stringheta, José Benício Paes Chaves,
among others, for providing the necessary support and viabilizing the participation of
Dr. Susanne Talcott in my thesis defense.
I am very grateful to the technical assistance of Valério Poleto and to the
friendship of my co-works of the Laboratory of Natural Pigments and Bioactive
Compounds at the Department of Food Technology, UFV, for providing a harmonious,
organized and pleasure work environment. Among other friends, I very much appreciate
Aline Arruda, Pollyanna, Paula, Erika, Neuma and Aline Nachtigall for the pleasure
shared hours during academic works. I want to thank a lot of friends including Manuela,
iii
Washington, Mirian, Roney, Aurelia, Taila, Danilo, Luciana, Flavia, Joesse, Roberta,
Alice, Leonardo, Meliza, Laura, Roberta, Rita, Fabiana, Jonson, Milton, Márcia,
Marília, Solange, Fernanda, Bruna, Mauricio, Igor, Alexandre, for their friendly
presence at the Department of Food Technology, UFV.
I really appreciate the receptivity, generous help in the lab and friendly presence
of my co-works Gabriela, Armando, Emily, Lisbeth Pacheco, Chris Duncan, Jorge
Cardona, Kim, Salvador, Michelle, Patricia, Luis Fernando, Warda, Keily at the
Department of Food Science and Nutrition and at Centeq Research Parkway,
Texas A&M University, USA. I would like to express my gratitude to Giuliana Norato
and Kimmy for the friendship and generous technical assistance with the experiment
analysis.
I am very grateful to the friends Paulo, Carmem, Frederico, Adolfo, Cristiane
and Helena for their friendship and help in USA.
I very much appreciate the technical support with the flow cytometry analysis of
Dr. Roger Smith, Texas A&M University, College Station, USA.
I highly value the collaborators of the Department of Food Science and
Technology, UFV, for their continuous support. Among names and nicknames: Adão,
Tineca, Juarez, Geralda, Vaninha, Sueli, Bilico, Lelé, Perereca, Luiz, Zé Geraldo, Maria
Rita, Divino, Piu, Pi. I really appreciate Gilcemir, Salvadora, Sueli and Geralda for their
help with CAPES process. They also did not measure efforts to viabilize Dr. Susanne
trip to Brazil. I also want to thank Alice for my English classes.
I want to express my gratitude to my family Liliane, Edson, Regina, Vicência,
José Antônio, Nayarah, Túlio, among others, for their support and care.
Last, but not least, I am very grateful to my lovely parents, Helena and Dirceu,
my husband Hernani Santana, and my son Vítor, for their active participation and
constant support. Their love, patience, dedication, care and friendship are the main
reason of my constant motivation.
I would express my sincere acknowledgments for all that contributed to my
personal growth and to the development of this work.
iv
BIOGRAPHICAL SKETCH
MICHELE CORRÊA BERTOLDI was born in September 12th, 1980, in Juiz de
Fora, Minas Gerais, Brazil. She earned the degree of Food Engineering through
Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil, in 2004. At the same
year, she was admitted by the Graduate Program in Food Science and Technology at the
Department of Food Technology of the Universidade Federal de Viçosa and received a
fellowship of National Counsel of Technological and Scientific Development (CNPq).
She completed her Master degree in Food Science and Technology with a minor in
Chemistry, Physics, Physical chemistry and Biochemistry of Food, in 2006. Following
that, she was admitted by the same Graduate Program at the Department of Food
Technology of the Universidade Federal de Viçosa, and her studies were funded by the
Foundation Coordination for the Improvement of Higher Education Personnel
(CAPES). She received a fellowship of this Governmental Foundation to conduct part
of her Doctoral studies at the Centeq Research Parkway and at the Department of Food
Science and Nutrition of Texas A&M University, College Station, USA (2008-2009). In
2009, she pursued the degree of Doctor of Science.
v
TABLE OF CONTENTS
LIST OF TABLES ...........................................................................................................ix
LIST OF FIGURES .........................................................................................................xi
RESUMO .......................................................................................................................xiv
ABCTRACT .................................................................................................................xvii
CHAPTER
1. INTRODUCTION ........................................................................................................ 1
Carcinogenesis ................................................................................................................. 1
Polyphenols in cancer prevention .................................................................................... 2
Bioactive effects of mango phytochemicals ................................................................... 3
Oxidative stress and cancer............................................................................................... 6
Bioavailability of polyphenols .......................................................................................... 7
Apoptosis ........................................................................................................................ 11
Extrinsic death receptor pathway.............................................................................. 11
Mitochondrial pathway .............................................................................................. 14
Cell cycle regulation ....................................................................................................... 15
Anticarcinogenic effects of polyphenols in normal cells as compared to cancer cells... 18
Objectives ....................................................................................................................... 19
References....................................................................................................................... 21
vi
2. ANTICARCINOGENIC EFFECTS OF POLYPHENOLS FROM DIFFERENT
MANGO (Mangifera indica L.) VARIETIES ................................................................ 37
Abstract ........................................................................................................................... 37
Introduction..................................................................................................................... 38
Material and Methods ..................................................................................................... 39
Chemicals............................................................................................................... 39
Plant material ......................................................................................................... 39
Extraction of polyphenols ...................................................................................... 40
Antioxidant capacity (ORAC assay)...................................................................... 41
HPLC-DAD and HPLC-ESI/MSn Analysis .......................................................... 42
Cell culture............................................................................................................. 43
Cell proliferation .................................................................................................... 43
Cell cycle kinetics .................................................................................................. 44
Quantitative RT-PCR............................................................................................. 44
Reactive oxygen species (ROS)............................................................................. 45
Statistical analysis.................................................................................................. 45
Results and Discussion ................................................................................................... 46
Total polyphenols and antioxidant activity of mango varieties ............................. 46
HPLC-DAD and HPLC-ESI/MSn analysis of Haden and Ataulfo polyphenols ... 47
Cell-growth supressive activity of Haden and Ataulfo mango polyphenols on
different cancer cell lines ................................................................................................ 49
Cell-growth supressive activity of mango polyphenols from different mango
varieties on colon cancer cells ........................................................................................ 51
Cell-growth supressive activity of ataulfo polyphenols on cancer cells as
compared to normal cells ................................................................................................ 54
Cell cycle regulation .............................................................................................. 55
Gene transcriptional regulation.............................................................................. 57
Protective effects against reactive oxygen species (ROS) ..................................... 60
Conclusion ...................................................................................................................... 62
References....................................................................................................................... 64
vii
3. ABSORPTION AND BIOLOGICAL ACTIVITIES OF POLYPHENOLS FROM
DIFFERENT MANGO (Mangifera indica L.) VARIETIES as affected by βGLUCOSIDASE hydrolysis .......................................................................................... 70
Abstract ........................................................................................................................... 70
Introduction..................................................................................................................... 72
Material and Methods ..................................................................................................... 73
Plant material ......................................................................................................... 73
Extraction of polyphenols ...................................................................................... 74
Enzymatic hydrolysis............................................................................................. 75
Fractionation of mango phenolic extracts.............................................................. 76
HPLC-DAD and HPLC-ESI/MSn Analysis .......................................................... 77
Antioxidant capacity (ORAC assay)...................................................................... 78
Cell culture............................................................................................................. 79
Cell proliferation .................................................................................................... 79
Transepithelial transport model ............................................................................. 80
Statistical analysis.................................................................................................. 81
Results and Discussion ................................................................................................... 81
Mango pulp phenolic content and antioxidant activity ......................................... 81
Enzymatic hydrolysis of mango pulp polyphenols ................................................ 82
Phenolic content and antioxidant capacity ...................................................... 82
HPLC-DAD and HPLC-ESI/MSn Analysis ................................................... 84
Transepithelial transport model ............................................................................. 89
Cell-growth supressive activity of mango pulp polyphenols................................. 93
Cell-growth supressive activity of low and high molecular weight polyphenolsrich fraction .................................................................................................................... 99
Conclusion .................................................................................................................... 105
References..................................................................................................................... 107
4. SUMMARY AND GENERAL CONCLUSIONS.................................................... 120
APENDIX ..................................................................................................................... 122
viii
LIST OF TABLES
Table 1:
Total polyphenols and antioxidant activity (ORAC) of different mango
varieties .................................................................................................. 46
Table 2:
Polyphenols profile from the mango varieties Ataulfo, Haden, Kent,
Francis, and Tommy Atkins determined by HPLC-DAD and HPLCESI-MSn analysis .................................................................................. 49
Table 3:
IC50 values of polyphenols extracted from Ataulfo and Haden mango
varieties for growth suppression of different human cancer cell lines . 50
Table 4:
IC50 values of polyphenols extracted from mango varieties for growth
suppression of human SW-480 colon cancer cells................................ 53
Table 5:
Total polyphenols and antioxidant activity (ORAC) of different mango
varieties ................................................................................................. 82
Table 6:
Total phenolic content and antioxidant activity of hydrolyzed and
control mango phenolic extracts ........................................................... 83
Table 7:
HPLC-DAD and HPLC-ESI-MSn of mango phenolic extracts (Control)
from different mango varieties.............................................................. 86
Table 8:
Effect of enzymatic hydrolysis on the phenolic acids profile of different
mango varieties. .................................................................................... 88
ix
Table 9:
Absorption (%) of phenolic acids through Caco-2 colon cancer cells
following 2h incubation with control and hydrolyzed extracts............. 91
Table 10:
Effect of the treatment with control and hydrolyzed mango phenolic
extracts on the growth suppression of MDA-MB-231 breast and HT-29
colon human cancer cell lines, expressed in terms of IC50 values (mg
GAE/L).................................................................................................. 95
Table 11:
Cell-growth suppressive effects of mango polyphenols (control) on
MDA-MB-231 breast and HT-29 colon human cancer cell lines,
expressed in terms of IC50 values (mg mango pulp/mL culture medium).
............................................................................................................... 95
Table 12:
Total phenolic content (TPC) and antioxidant activity of the hydrolyzed
phenolic extract and its fractions ....................................................... 102
Table 13:
Effect of the treatment with HMW (F1) and LMW (F2) fractions on the
growth suppression of MDA-MB-231 breast and HT-29 colon human
cancer cell lines, expressed in terms of IC50 values (mg GAE/L) ..... 100
x
LIST OF FIGURES
Figure 1:
Chemical structure of 1,2,3,4,6-penta-O-galloyl-β-D-glucose (PGG), the
precursor of gallotannins......................................................................... 4
Figure 2:
Routes of absorption and metabolism of dietary polyphenols in human
body......................................................................................................... 7
Figure 3:
Pathways involved in apoptosis signaling: Fas-mediated extrinsic death
receptor pathway and mitochondrial pathway; (
) inhibition; (
)
activation............................................................................................... 13
Figure 4:
Procedure used for extraction of mango polyphenols........................... 41
Figure 5:
Representative chromatograms at 280 nm of Ataulfo (A) and Haden
polyphenols (B). (1) gallotannins; (2) gallic acid. Representative
chromatograms at 366 nm of Ataulfo (A1) and Haden (B1) (3)
mangiferin ............................................................................................ 48
Figure 6:
Cell proliferation of prostate LnCap, leukemia Molt-4, lung A-549,
breast MDA-MB-231 and colon SW-480 cancer cells treated with
Ataulfo (A) and Haden (B) polyphenols. Cells were treated with
different concentrations of extracts and cell growth was assessed after
72 h incubation. Values are means ± SD, n=3 ...................................... 50
Figure 7:
Cell-growth suppressive effects of polyphenols from Kent, Tommy
Atkins, Haden, Ataulfo and Francis mango varieties on the colon SW480 human cancer cells. Cells were incubated with extracts and net
xi
growth was measured after 72 h incubation. Values are mean ± SD,
n=3.. ...................................................................................................... 52
Figure 8:
Cell-growth suppressive effects of Ataulfo polyphenols on the human
SW-480 colon cancer cells and colonic myofibroblasts CCD-18Co cells.
Cells were incubated with extracts and net growth was measured after
48 h. Values are mean ± SD, n=3. Asterisk indicates a significant
difference compared to untreated control (*) p ≤ 0.01 ......................... 55
Figure 9:
Cell cycle analysis of SW-480 colon cancer cells teated with Ataulfo
and Haden polyphenols for 24 h. Values are means ± SE (n=3), different
letters indicate significance at p < 0.05................................................. 56
Figure 10:
mRNA expression of SW-480 colon cancer cells treated with Ataulfo
(A) and Haden (B) polyphenols after 24 h and analyzed by real time
PCR as ratio to TATA-binding protein (TBP) mRNA. Values are means
± SE (n=3). Different letters indicate significance at p < 0.05. ............ 58
Figure 11:
Protective effects of Ataulfo and Haden polyphenols against H2O2induced ROS production on SW-480 cancer cells (A) and CCD-18Co
non-cancer cells (B). Cells were pretreated with extract for 24h and
exposed to 200μM H2O2 for 2h. Values are means ± SD (n=6),
different letters indicate significance at p < 0.05.. ................................ 62
Figure 12:
Procedure used for extraction of mango polyphenols........................... 75
Figure 13:
Procedure used for fractionation of mango phenolic extract. ............... 77
Figure 14:
Representative chromatograms at 280 nm (A) and 340 nm (B) of
Ataulfo mango pulp polyphenols from control phenolic extracts. Peak
assignments: (1) gallic acid; (2) p-OH-benzoic acid; (3) gallotanin; (4)
vanillic acid; (5) caffeic acid; (6) mangiferin; (7) p-coumaric acid; (8)
ferulic acid............................................................................................. 85
xii
Figure 15:
Representative chromatograms at 280 nm (A) and 340 nm (B) of
Ataulfo mango pulp polyphenols from hydrolysed phenolic extracts.
Peak assignments: (1) gallic acid; (2) p-OH-benzoic acid; (3)
gallotannin; (4) vanillic acid; (5) caffeic acid; (6) mangiferin; (7) pcoumaric acid; (8) ferulic acid. ............................................................. 87
Figure 16:
Chromatograms (280 nm) from samples taken from the basolateral
compartment of Caco-2 cells following 2h of incubation with HBSS (pH
6.0) (A), control (B) and hydrolyzed (C) Ataulfo phenolic extracts. Peak
assignments: (1) gallic acid; (2) p-OH-benzoic acid; (3) vanillic acid; (4)
caffeic acid; (5) p-coumaric acid; (6) ferulic acid................................. 90
Figure 17:
Cell-growth suppressive effects of mango pulp polyphenols on estrogen
independent MDA-MB-231 breast (A) and HT-29 colon (B) human
cancer cells. Cells were incubated for 48h with control (c) and
hydrolyzed (h) phenolic extracts. Values are mean ± SD, n=3............. 94
Figure 18:
Chromatograms at 280 nm from HMW (A) and LMW (B) fractions
from Ataulfo hydrolyzed extract. Peak assignments: (1) gallic acid; (2)
p-OH-benzoic acid; (3) vanillic acid; (4) caffeic acid; (5) mangiferin; (6)
p-coumaric acid; (7) ferulic acid........................................................... 99
Figure 19:
Cell-growth suppressive effects of mango pulp polyphenols on estrogen
independent MDA-MB-231 breast (A) and human HT-29 colon (B)
cancer cells. Cells were incubated for 48h with HMW and LMW
fractions from Ataulfo hydrolyzed extract. Error bars represent the
standard error of the mean (n=3)......................................................... 101
Figure 20:
Effect of the treatment with equal dilutions of the HMW (F1) and LMW
fractions from Ataulfo hydrolyzed extracts on the growth suppression of
MDA-MB-231 breast and HT-29 colon human cancer cell lines,
expressed in percentage of control. Error bars represent the standard
error of the mean (n=3). ...................................................................... 102
xiii
RESUMO
BERTOLDI, Michele Corrêa, D.Sc.,Universidade Federal de Viçosa, dezembro, 2009.
Capacidade antioxidante, efeitos anticarcinogênicos e absorção de polifenóis de
de manga (Mangifera indica L.) in vitro. Orientador: Paulo Cesar Stringheta. Coorientadores: Susanne U. Mertens-Talcott e Tânia Toledo de Oliveira.
Polifenóis presentes em polpa de manga, incluindo galotaninos, glicosídeos de
flavonóides, ácido gálico, derivados da benzofenona, e mangiferina, têm demonstrado
propriedades anticarcinogênicas. A etapa de deglicosilação por β-glicosidases tem se
mostrado necessária ao metabolismo e à absorção de polifenóis derivados da dieta pelo
organismo humano, o que poderia influenciar suas propriedades anticarcinogênicas. O
objetivo deste estudo foi elucidar os efeitos anticarcinogênicos de polifenóis extraídos
da polpa de manga de diferentes variedades (Francis, Kent, Ataulfo, Tommy Atkins e
Haden) em diferentes tipos de câncer. Os efeitos antiproliferativos de polifenóis de
manga foram estudados utilizando modelos in vitro de cultura de células cancerosas,
incluindo as linhagens celulares de câncer humano Molt-4 (leucemia), A-549 (câncer de
pulmao), MDA-MB-231 (câncer de mama), LnCap (câncer de próstata), SW-480
(câncer de colón) e células de colón não cancerosas CCD-18Co. Os mecanismos
moleculares envolvidos nas propriedades anticarcinogênicas de polifenóis de manga
foram investigados. O efeito do tratamento com polifenóis na expressão gênica, na
regulação do ciclo celular e na produção de espécies reativas de oxigênio em células
cancerosas de colón humano SW-480 foi investigado por RT-PCR, citometria de fluxo
e quantificação da intensidade de fluorescência, respectivamente. Além disso, o efeito
da hidrólise de polifenóis de manga pela enzima β-glicosidase na atividade antioxidante,
na supressão do crescimento tumoral e na absorção intestinal in vitro através da
monocamada de células de adenocarcinoma de colón humano Caco-2 foi avaliado.
Ademais, o efeito antiproliferativo de frações fenólicas enriquecidas com polifenóis de
baixo e elevado peso molecular em células cancerosas de colón (SW-480) e mama
(MDA-MB-231) foi estudado. Células cancerosas foram tratadas com extratos fenólicos
das variedades Ataulfo e Haden, as quais foram selecionadas em razão da maior
capacidade antioxidante quando comparada a outras variedades. Polifenóis de Ataulfo e
Haden inibiram o crescimento de todas as linhagens celulares. SW-480 (câncer de
cólon), MOLT-4 (leucemia) e MDA-MB-231 (câncer de mama) apresentaram
xiv
igualmente maior sensibilidade ao tratamento com polifenóis de Ataulfo, enquanto SW480 e MOLT-4 mostraram-se mais sensíveis ao tratamento com polifenóis de Haden,
segundo resultados obtidos por contagem de células. O efeito antiproliferativo dos
extratos fenólicos de todas as variedades de manga foi avaliado em células cancerosas
de colón humano (SW-480). As variedades Ataulfo e Haden demonstraram maior efeito
supressor, seguidas de Kent, Francis e Tommy Atkins. Quando células cancerosas de
colón SW-480 foram tratadas com 5 mg GAE/L de polifenóis de Ataulfo, o crescimento
celular foi inibido em ~79%, enquanto a proliferação de miofibroblastos não cancerosos
CCD-18Co não foi inibida. A supressão do crescimento celular pelo tratamento com
polifenóis de Ataulfo e Haden em células de câncer de colón SW-480 foi associada com
o aumento na expressão gênica de biomarcadores de apoptose (caspase 8, Bax e Bim) e
reguladores do ciclo celular (PKMYT1), atraso do ciclo celular e alteração na produção
de espécies reativas de oxigênio. Os extratos fenólicos da polpa de manga continham
ácido gálico, mangiferina, derivados de ácidos fenólicos e galotaninos, os quais foram
caracterizados por análises em HPLC-DAD e HPLC-ESI-MSn antes e após a hidrólise
enzimática (0.17 mg β-glicosidase 1000 KU/g polpa de manga / 4 h / 35°C). Ácidos
fenólicos incluindo ácido gálico, caféico, ferúlico, p-coumárico e p-hidroxibenzóico
consistiram os principais compostos derivados da hidrólise enzimática. Monocamadas
de células Caco-2 foram incubadas por 2h no compartimento apical com extratos
controle e hidrolisado. Quando incubadas com o extrato hidrolisado, ácido gálico,
caféico, ferúlico, p-coumárico, vanílico e p-hidroxibenzóico foram detectados no
compartimento basolateral, enquanto apenas ácido gálico foi detectado quando as
células foram tratadas com o extrato controle. Polifenóis de elevado peso molecular,
incluindo mangiferina e galotaninos, não foram transportados. Polifenóis de polpa
(controle) de todas as variedades inibiram a proliferação de células humanas de câncer
de colón HT-29 (0-27 μg de ácido gálico equiv./mL) e de mama MDA-MB-231 (0-24
μg GAE/mL) em até 99.8 e 89.9 %, respectivamente. Apesar da hidrólise enzimática ter
aumentado a absorção de polifenóis de manga, não houve aumento significativo na
atividade antioxidante, no conteúdo fenólico e na supressão do crescimento de células
cancerosas de colón e mama. Ademais, ambas as frações fenólicas enriquecidas com
polifenóis de baixo (138-194 Da) e elevado (422; 788-1852 Da) peso molecular
inibiram o crescimento de células cancerosas de colón e mama na mesma extensão (0-
xv
20 μg GAE/mL), o que poderia indicar que a atividade anticarcinogênica de polifenóis
de manga seria independente da hidrólise enzimática. Estes resultados corroboram
resultados obtidos em estudos in vivo, que sugerem que a grande parte dos polifenóis
não seriam absorvidos em sua forma intacta através do intestino delgado, mas poderiam
ser hidrolisados por enzimas intestinais em ácidos aromáticos de baixo peso molecular,
os quais seriam posteriormente absorvidos; ou ainda, quando não absorvidos, poderiam
alcançar o intestino grosso, modulando a microflora intestinal e, desta forma,
contribuiriam para reduzir o risco de câncer de cólon. Desta forma, polifenóis de polpa
de manga de diferentes variedades exibiram efeitos anticarcinogênicos em modelos de
cultura de células, os quais poderiam não ser necessariamente dependentes da hidrólise
enzimática pela β-glicosidase.
xvi
ABSTRACT
BERTOLDI, Michele Corrêa, D.Sc.,Universidade Federal de Viçosa, December, 2009.
Antioxidant capacity, anticancer effects and absorption of mango (Mangifera
indica L.) polyphenols in vitro. Adviser: Paulo Cesar Stringheta. Co-advisers:
Susanne U. Mertens-Talcott and Tânia Toledo de Oliveira.
Polyphenols found in mango pulp, including gallotannins, flavonol glycosides,
gallic acid, benzophenone derivatives and mangiferin have shown anticancer activity.
Biological activities of polyphenols have been related to their bioavailability.
Deglycosylation by β-glucosidases is a critical step in the metabolism and absorption of
dietary polyphenols in humans, which might influence their anticancer properties. The
objective of this study was to elucidate the anti-cancer effects of mango polyphenols of
several varieties (Francis, Kent, Ataulfo, Tommy Atkins and Haden) in different types
of cancer. The antiproliferative effects of mango polyphenols were studied in vitro
using different cancer cell lines including Molt-4 leukemia, A-549 lung, MDA-MB-231
breast, LnCap prostate, SW-480 colon cancer cells and the non-cancer colon cell line
CCD-18Co. Molecular mechanisms involved on the anti-cancer activities of mango
polyphenols were assessed. The effect of mango polyphenols on gene expression, cell
cycle regulation and reactive oxygen species production on colon cancer cells SW-480
were investigated by RT-PCR, flow cytometry and fluorescence intensity measurement,
respectively. The effect of the hydrolysis of mango polyphenols with β-glucosidase on
their antioxidant activity, cancer cell-growth suppression activity and in vitro intestinal
absorption through human colon adenocarcinoma Caco-2 cell monolayers was
evaluated. In addition, the antiproliferative effect of high and low molecular weight
polyphenols rich-fractions on colon (SW-480) and breast (MDA-MB-231) cancer cells
was studied. Cell lines were incubated with Ataulfo and Haden phenolic extracts, which
were selected based on their superior antioxidant capacity compared to the other
varieties. Ataulfo and Haden polyphenols inhibited the growth of all human cancer cell
lines. SW-480 (colon cancer), MOLT-4 (leukemia) and MDA-MB-231 (breast-cancer)
were statiscally equally most sensitive to Ataulfo, whereas SW-480 and MOLT-4 were
the most sensitive cell lines to Haden, as determined by cell counting. The efficacy of
phenolic extracts from all mango varieties in inhibiting cell growth was tested on SW480 colon carcinoma cells. Ataulfo and Haden demonstrated superior efficacy, followed
xvii
by Kent, Francis and Tommy Atkins. At 5 mg GAE/L, Ataulfo inhibited the growth of
colon SW-480 cancer cells by ~79% while the growth of non-cancer colonic
myofibroblasts CCD-18Co cells was not inhibited. The growth inhibition exerted by
Ataulfo and Haden polyphenols on SW-480 cells was associated with an increased
mRNA expression of pro-apoptotic biomarkers (caspase 8, Bax and Bim) and cell cycle
regulators (PKMYT1), cell cycle arrest and an alteration in the generation of reactive
oxygen species. Phenolic extracts from mango pulp contained gallic acid, mangiferin,
phenolic acid derivatives and gallotannins, which were characterized by HPLC-DAD
and HPLC-ESI-MSn analysis before and after enzymatic hydrolysis (0.17 mg βglucosidase 1000 KU/g mango pulp/ 4 h / 35°C). Phenolic acids including gallic,
caffeic, ferulic, p-coumaric and p-hydroxybenzoic acids consisted the main compounds
derived from enzymatic hydrolysis. Caco-2 cell monolayers were incubated for 2h on
the apical side with hydrolyzed and non-hydrolyzed mango extracts. Gallic, caffeic,
ferulic, p-coumaric, vanillic and p-hydroxybenzoic acids were detected on the
basolateral side for hydrolyzed extract but only gallic acid was detected for the nonhydrolyzed extract. High molecular weight polyphenols, mangiferin and gallotannins,
were not transported. Mango pulp polyphenols (control) from all varieties inhibited the
proliferation of HT-29 colon (0-27 μg of gallic acid equiv/mL) and MDA-MB-231
breast (0-24 μg GAE/ mL) human cancer cells by up to 99.8 and 89.9 %, respectively.
Despite enhanced absorption facilitated by enzymatic hydrolysis, a significant increase
in antioxidant activity, phenolic content and antiproliferative effects on breast and colon
cancer cells was not observed. Additionally, both high (422; 788-1852 Da) and low
(138-194 Da) molecular weight polyphenols rich-fractions equally inhibited cell
proliferation of colon and breast cancer cells at the same extent (0-20 μg of gallic acid
equiv/mL), which may indicate that the anti-cancer efficacy of mango polyphenolics is
not dependent on enzymatic hydrolysis. These results corroborate previous findings
from in vivo studies, which suggest that the most of mango polyphenols are not
absorbed intact through the small intestine, but may be hydrolyzed by intestinal
enzymes into low molecular weight aromatic acids, which would be later absorbed; or
when polyphenols are not absorbed, they likely reach the large intestine, modulating the
gut microflora, and thus they contribute to reduce the risk of colon carcinogenesis.
xviii
Overall, polyphenols from several mango varieties exerted anti-cancer effects, and these
effects may not require enzymatic hydrolysis by β-glucosidase.
xix
INTRODUCTION
Carcinogenesis
Cancer is one of the leading chronic diseases and causes of death worldwide (1).
In 2008, 12 million of new cases of cancer and around 7 million of deaths were
estimated, with the highest incidence for lung, breast and colon cancer (1). Colon cancer
is the third most common cancer in USA for both men and women, with 108,070
estimated cases of colon and 40,740 cases of rectal cancer diagnosed in 2008 (2).
According to INCA (National Institute of Cancer) (3), the occurrence of 489.270 new
cases of cancer is expected to 2010-2011 in Brazil, with the highest incidence for skin
(114,000 cases), prostate (52,000), breast (18,000) and colon (28,000).
Neoplasia consists in an abnormal, uncontrolled and exaggerated proliferation of
cells as a result of damage in mechanisms of cell cycle regulation or alteration in genes
that regulate the growth and differentiation of cells, which may be benign or malignant
(4-5). Cancer is a term used for diseases in which abnormal cells divide without control
and are able to spread through the blood and lymph systems and invade others tissues in
the body. The process is initially characterized in cell mutation (initiation) as a result of
a change in the genetic material of the cell primes. This alteration may occur
spontaneously or by an agent that causes cancer (carcinogen). Development of cancer
includes invasion, which refers to the direct migration and penetration by cancer cells
into neighboring tissues, and metastasis, which is the ability of cancer cells to penetrate
into lymphatic and blood vessels, circulate through the bloodstream, and then invade
normal tissues elsewhere in the body. Cancerous cells present uncontrolled growth,
evasion of apoptosis, self-sufficiency in growth signals, insensitivity to growthinhibitory signals, limitless replicative potential, sustained angiogenesis, invasion of
adjacent tissues, and sometimes metastasis, process by which cells spread to other
locations in the body via lymph or blood. These malignant properties are hallmarks of
cancer and differentiate cancer cells from begin tumors, which are self-limited, and do
not invade or metastasize (6-10).
The inadequate functioning of genes which regulate the proliferation of
transformed cells is a major factor in neoplasia. Cancer formation is a multi-step
1
process that may involve the sequential activation of oncogenes as well as the
inactivation of tumor suppressor genes often in the same clone of cells. These genetic
changes cause phenotypic alterations in tumor cells that allow them to continue to
survive and expand. Both tumor suppressor genes and oncogenes are responsible for
proliferation control or differentiation after mutation, which may result in an
overexpression of proteins and tumor formation (11). In normal cells, oncogenes often
are underexpressed or inactivated (12). Cancer cells are generated, when DNA is
damaged, and cells do not undergo apoptosis but instead oncogenes are activated to
prevent cell death, resulting in continued and disorderly proliferation (11). Tumor
suppressor genes (anti-oncogenes) normally function to limit cell proliferation. Their
loss of function d facilitates cancer development, usually in combination with other
genetic changes (13). Tumor suppressor genes, or more precisely, the proteins they
encode, either have a dampening or repressive effect on the regulation of the cell cycle
or promote apoptosis, and sometimes both. The products of oncogenes and tumor
suppressor genes have therefore become an important target for new anti-cancer drugs.
Thus, knowledge regarding the cancer molecular biology, following the identification
and functional characterization of many oncogenes and tumor suppressor genes are
critical for therapeutic approach in targeting and eliminating cancer cells (11, 14).
Cell culture models have been extensively used as in vitro trials to evaluate the
biological activities of several drugs and natural compounds, including polyphenols
(15-19). Because the great availability of different types of human cancer cell lines,
including colon, prostate, breast, and so on (20), the use of cell culture models permits
evaluation of the potential anticancer effects with higher specificity, reduced cost, and
in a shorter period of time.
Polyphenols in cancer prevention
Clinical and epidemiological studies have been related the fruit and vegetable
consumption to the reduced risks of several chronic diseases, including cancer.
Therefore, the risk of mortality associated with cancer might be reduced by diet
modification (21-23). The health benefits ascribed to fruit and vegetables are mainly
related to their bioactive compounds, which includes vitamins C and E, tocopherol,
2
beta-carotene, and phenolic compounds, which comprise the major antioxidant
compounds derived from diet (24).
Phenolic compounds include a complex mixture of secondary plant metabolites,
with high diversity in chemical structure and reactivity. Chemically, they are composed
by aromatic rings with one or more hydroxyl groups, including their functional
derivatives (25). Currently, more than 8000 polyphenols have been identified (17),
including phenolic acids, flavonoids, lignans, estilbens, cumarins and tannins, and their
consumption has been estimated in 1 g/day (26).
Polyphenols are associated not only with color and sensory properties, but also
are increasingly being considered as natural cancer chemopreventive compounds based
on safety and efficacy assessments (27-29). These compounds may prevent cancer by
interfering in enzymatic expression (30), improving intestinal health (31), reducing
oxidative stress (32-33), interfering in synthesis and repair of DNA (34), as well as
inducing apoptosis (35).
Although many studies have been shown in vitro and in vivo cancer-inhibitory
activities from individual compounds, the combination of a variety of phytochemicals in
fruits and vegetables may be increased in cancer prevention due to synergistic effects
(16, 35-36). Therefore, studies with complex mixtures of these compounds might
represent the health-benefits derived from food comsumption.
Bioactive effects of mango phytochemicals
Mangoes (Mangifera indica L.) are amongthe most important tropical fruits
marked in the world, with a global production exceeding 33 million tons in 2007.
Moreover, mangoes consist one of the most consumed fruits in Brazil due to sensorial
characteristics and nutritional value, ranking the seventh position worldwide in terms of
production in 2007 (37).
Consumer interest in mango fruits (Mangifera indica) and derived products has
been increasing in recent years based on fruit olfactory properties, attractive color,
nutritional content, and health-promoting phytochemicals, which confer them great
potential in preventing chronic diseases (38). The consumption of mangoes in Brazil is
characteristic of high-volume producer countries, and has been estimated to be 2.8
3
kg/person/year (39). Furthermore, there has been increasing interest in understanding
the mango phytochemicals biological properties due to their health-promoting
characteristics
such
as
antioxidant,
antitumoral,
anti-inflammatory,
and
immunomodulatory activities (40-45). Recently, mango fruit has been listed as a
nutrient-rich fruit into the unofficial classification and so-called superfruit due to its
considerable content of carotenoids, specially β-carotene, vitamin C and polyphenols,
including gallic acid, gallotannins, quercetin and kaempferol glycosides as well as
xanthone-C-glucosides (46-48). Gallotannins (Figure 1) represent the major high
molecular weight polyphenols found in mango pulp, with molecular weights ranging
from 332 (mono-O-galloyl-glucose) to over 1852 Da (undeca-O-galloyl-glucose) (47).
Figure 1. Chemical structure of 1,2,3,4,6-penta-O-galloyl-β-D-glucose (PGG), the
precursor of gallotannins.
In traditional medicine, the use of mango extracts as herbal drugs is widespread.
Vimang, a mango stem bark extract, have been used at least 10 years in Cuban medicine
with effectiveness against several diseases, like cancer (45). Moreover, studies in vitro
and in vivo performed with individual polyphenols found in mangoes have been shown
antitumor activities, such as induction of apoptosis, inhibition of tumor growth and
angiogenesis (49-54). Mangiferin has shown radioprotective (55-56), antioxidant,
antiviral, anti-inflammatory (57) and anticarcinogenic effects (58). This xanthone
4
demonstrated chemopreventive action against lung carcinogenesis induced by
benzo(a)pyrene in Swiss albino mice (59), which might be related to induction of
mitochondria permeability (60). Lupeol and related terpenoids exerted varied
cytotoxicity against several cancer cell types, including B16 2F2 mouse melanoma,
leukemia HL60, U937, K562 melanoma G36, human lung carcinoma (A-549) and
human colon adenocarcinoma (DLD-1) (61). Gallic acid showed to inhibit human
cancer cell lines including esophageal, gastric, breast, cervix and colon cancer cells HT29, Colo201 and colon 26 (mouse colon cancer). In esophageal cancer cells, apoptosisrelated molecular mechanism included up-regulation of the pro-apoptotic protein Bax,
PARP cleavage induction and caspase-cascade activity, down-regulation
of
antiapoptotic proteins Bcl-2 and Xiap and the survival Akt/mTOR pathway (62). The
effect of gallic acid and its alkyl esters is known to induce cell death or cell cycle arrest
in a variety of cancer cells, including colon cancer (63). This phenolic acid (100μM)
significantly increased the number of cells in G2/M phase, with a consequent reduction
of cells in G1 and S phases on human colon adenocarcinoma cells (Caco2) (64).
Additionally, several studies have been shown the anticancer-related activities of
gallotannins. 1,2,3,4,6-penta-O-galloyl-beta-d-glucose (PGG), which is the precursor of
gallotannins, showed to induce cell cycle arrest, cell proliferation and apoptosis in a cell
type-dependent manner (51-53), and to suppress tumor growth in vivo via angiogenesis
inhibition and stimulation of apoptosis (65).
Recently, studies performed with mango pulp extracts have been demonstrated
their chemopreventive potential (66-70), although conclusion regarding the biological
activities of the mango phenolic fraction is still not so clear (67-68), since other
bioactive compounds like L-ascorbic acid and carotenoids have been conjunctly
evaluated (66-68). Studies performed with mango extracts have suggested cell line
specificity; e.g. mango extracts have been shown to protect against prostate cancer in in
vitro and in vivo models (69) and to inhibit the cell cycle in the G(0)/G(1) phase of HL60 cells (66), although their lack of effectiveness against MCF-7 breast cancer cells has
also been reported (67). Moreover, anticancer effects of mango phytochemicals may
vary according to the variety as a result of changes in mango composition. It was
demonstrated the superior phenolic content and antioxidant activity in vitro of mango
pulp Ubá as compared to the varieties Palmer, Haden and Tommy Atkins (71).
5
The overall anti-cancer potential of mango polyphenols might be in part
associated to their antioxidant properties (72), which may contribute to enhance cell
defense capacity and modulate molecular pathways in target cells. These compounds
might also inhibit promotion and progression stages of cancer by interfering in cell
cycle regulation, signal transduction pathways, transcription and activating apoptosis
(activation of pro-apoptotic genes and pro-apoptotic proteins) in neoplastic cells (73).
Oxidative stress and cancer
Free radicals are highly reactive chemical species which contains one or more
unpaired electrons. These species comprise reactive oxygen species (ROS) and reactive
nitrogen species (RNS), which may attack molecules in vivo including proteins, lipids
and DNA (74). Superoxide, hydroxyl radical, hydrogen peroxide, hypochlorite and
singlet oxygen includes the most important reactive oxygen species (75), whereas
reactive nitrogen species constitute the molecules derived from nitric oxide (76).
Reactive oxygen species are produced as natural byproducts of oxygen reduction
during the normal metabolism and play an important role in cell signaling (77-78).
However, during times of environmental oxidative stress, ROS may cumulated and
result in significant damage to cell structures (DNA, RNA and proteins), which is
caused by an imbalance between the production of ROS and a biological system's
ability to readily detoxify the reactive intermediates or repair the resulting damage.
ROS can potentially cause DNA damage and promote tumor progression. Thus,
oxidative stress has been considered a hallmark of many tumors (79). In addition to
causing genomic instability, ROS are know to increase tumorigenesis by activating
signaling pathways that regulate cellular proliferation, angiogenesis, and metastasis (78,
80-81). Low or transient levels of ROS can activate cellular proliferation or survival
signaling pathways, while high levels of ROS can initiate damage or cell death (78).
Polyphenols are known to present anticancer activity due to their ability to
modulate expression/activity of antioxidative and phase II drug-metabolizing enzymes
as well as scavenging free radicals (82-84). In cancer cells, these bioactive compounds
may induce cell cycle arrest and apoptosis by causing generation of ROS to trigger
6
signal transduction (72). Interestingly, the induction in ROS generation by some dietary
anticancer agents is tumor cell specific, which may not occur in normal cells (85-87).
Bioavailability of polyphenols
Polyphenols are extensively metabolized either in tissues, once they are
absorbed by small intestine, or by the colonic microflora, that transform the nonabsorbed fraction or the fraction re-excreted in the bile (Figure 2). Once absorbed,
polyphenols are conjugated to form O-glucuronides, sulphate esters and O-methyl ether
in the small intestine and later in the liver, and no free aglycones are found in the
plasma. Conjugation (methylation, sulfation and glucuronidation) reduces polyphenols
potential toxicity, enhances their hidrophilicity and facilitates their biliary and urinary
elimination. Trough enterophatic recirculation, conjugated compounds are excreted by
the liver as components of bile into the intestine, while deconjugated compounds are
transformed by microbial enzymes before being absorbed (31, 88-90).
Tissues
Polyphenols
CP450
Liver
Small Intestine
Kidney
Intestinal enzymes
Bile
Large Intestine
(Colon)
Gut Microflora
Urine
Feces
Excretion
Figure 2. Routes of absorption and metabolism of dietary polyphenols in human body.
Most of polyphenols are found in food in the form of esters, glycosides, or
polymers that cannot directly be absorbed in their intact form through small intestine.
7
Thus, exogenous (luminal) deglycosylation by several β-glucosidases in small intestine,
or coordination between epithelial transportes and intracellular β-glucosidases facilitate
the absorption of some phenolic compounds such as flavonoid glycosides (90-91).
Because only aglycones and some glucosides can be absorbed in the small intestine,
most of dietary polyphenols need to be firstly transformed in the colon by intestinal
microflora before their metabolites may be absorbed (31, 88). Thus, gut bacteria can
hydrolyze glycosides, glucuronides, sulfates, amides, esters and lactones. They also are
responsible for reduction, descarboxilation, ring-cleavage, demethylation, and
dehydroxylation reactions. However, when the microflora is involved, the efficiency of
absorption is often reduced because the flora also degrades the aglycones into simple
aromatic acids. In addition, polyphenols, which present antimicrobial activities, can
interact specifically in a structure-dependent way with certain types of microorganisms
in the gut, also modulating the microbial population of the gastrointestinal tract. This
has effects on gastrointestinal health as well as in polyphenolic metabolism (92-93).
Therefore, the biotransformation by gut bacteria may result in metabolites with higher
or lower biological activity than the parent compounds (94-95).
Structural parameters of polyphenols including molecular weight, glycosylation
and esterification have a major impact on intestinal absorption (96-97). According to
the Lipinski`s Rule of 5 (98), compounds with five or more hydrogen bond donors (OH
and NH groups), ten or more hydrogen bond acceptors (N and O), molecular weight
higher than 500, and Log P greater than 5 are usually poorly absorbed following oral
administration. In general, low molecular weight phenolics such as caffeic acid are
directly absorbed through the gut barrier by passive diffusion, whereas high molecular
weight polyphenols such as flavonoids and proanthocyanidins are typically transformed
in the colon by intestinal microflora into a wide array of bioactive low molecular weight
metabolites, which are later absorbed through the colonic barrier (90, 99). Thus, the
enzyme and microbial-mediated biotransformation and active efflux are factors that also
limit the availability of polyphenols. Because of their unfavorable structure as substrates
for the cytochrome P450s, most polyphenolics are not subject to phase I metabolism
and, therefore, these compounds can directly undergo methylation, glucuronidation and
sulfation steps during phase II metabolism (90).
8
In addition to molecular weight, glycosylation has a considerable impact in the
intestinal absorption of dietary polyphenols, because it influences chemical, physical
and biological properties of polyphenols and it directly affects its polarity and partition
coefficient (26). The aglycones are able to be absorbed by the small intestine. Thus, a
commonly accepted concept regarding polyphenols intestinal absorption is that
glycosylated polyphenols need to be converted to the aglycone by glycosidases in the
food or gastrointestinal mucosa, or from the colon microflora in order to be absorbed by
passive diffusion (100). Esterification has also an impact in polyphenols absorption.
Esterified polyphenols are, in general, lower absorbed than their non-esterified forms.
Galloylated catechins, for example, were better recovered in human urine after black tea
consumption than non-galloylated catechins, demonstrating their low availability (101).
Therefore, as a result of structural impediment and molecular size, high
molecular weight compounds like tannins and flavonoids are not absorbed through
intestinal gut barrier, but hydrolyzed by gut microflora (26, 31). It has been confirmed
by several in vitro and in vivo studies. As for the smaller oligomers, it was reported that
radiolabeled monomers, dimers, and trimers can be transported across in vitro cell layer
of Caco-2 cells, whereas the larger oligomers adhere to the cell surface (102-103).
Deprez et al. (2001) (103) demonstrated that proanthocyanidins dimers, trimers and
polymers were hardly absorbed through the human colon carcinoma (Caco-2)
monolayer at higher transepithelial electric resistance (TEER values= 509 Ω.cm2), and
absorption of proanthocyanidins polymers with an average polymerization degree of 6
(molecular weight 1,740) was about 10-fold lower. According Holt et al. (2002) (104),
dimeric procyanidins were detected in human plasma as early as 30 min after the
consumption of a cocoa, whereas no evidence of absorption of oligomeric procyanidins
was found. Similarly, Prasain et al. (2009) (105) showed that monomeric catechins and
their methylated metabolites, as well as proanthocyanidins up to trimers were detected
in blood samples after oral consumption of grape seed proanthocyanidin-rich extracts.
Although the absorption of procyanidin dimmers has been demonstrated using animal
models (106), Appeldoorn et al. (2009) (107) showed that these compounds are also
metabolized by human microbiota. Interestingly, even though punicalagin was detected
in plasma of rats after oral ingestion and it is known to be the highest molecular weight
compound (1084 Da) ever absorbed in animals so far (108), studies showed that this
9
water soluble ellagitannin found in pomegranates was hydrolyzed into ellagic acid in
vitro across the mitochondrial membrane (109), and converted in vivo by gut microflora
to 3,8-dihydroxy-6H-dibenzo[b,d]pyran-6-one (urolithin A, UA) derivatives (18).
Caco-2 cell monolayers are among the most functional in vitro models in the
field of drug absorption and permeability, which has been used to predict the in vivo
intestinal absorption and transport of various polyphenols including phenolic acids
(110), flavonoids (111), and procyanidins (103). Several studies have been shown that
β-glucosidase activity is related to the intestinal absorption of glycosides in vivo and in
caco-2 cell monolayers (112-117). The β-glucosidases (β-glucan glucohydrolase; EC
3.2.1.21) are a widespread group of enzymes that hydrolyze a broad variety of
glycosides including aryl- and alkyl- β -D-glycosides, where β-D-galactoside and β-Dglucoside substrates can be hydrolyzed with comparable efficiencies (118). In
mammals, several β-glucosidases have been characterized, including the lysosomal β glucosidase (also called acid β-glucosidase or glucocerebrosidase), lactase phlorizin
hydrolase (LPH) and cytosolic (or broad-specificity) β-glucosidase (118). The human
cytosolic β-glucosidase (hCBG) constitutes a group of nine enzymes with similarities in
the amino-acid and structural features related to their substrate specificities. They are
found in the cytosol of liver, spleen, kidney, small intestine, and lymphocytes of
mammals and can catalyze the hydrolysis of O-linked β-glycosidic bonds at the nonreducing end of carbohydrates with retention of anomeric configuration (118).
Gallotannins represent the major compounds found in mango pulp, with
molecular weight ranging from 332 (mono-O-galloyl-glucose) to up to over 1852 Da
(undeca-O-galloyl-glucose) or large (47). Considering that punicalagin (1084 Da) was
reported to be the highest molecular weight compound absorbed intact through the gut
barrier after oral ingestion (108), it is assumed that higher molecular weight
polyphenols would not be absorbed intact through human gut. However, limited
bioavailability of these compounds might not limit their biological properties, since the
highest local concentration of these compounds is found in the gut lumen, where
polyphenols may be protective against colon carcinogenesis (26, 31, 103). Therefore, it
is relevant to measure the biological activity of polyphenols on cultured cells or isolated
tissues in their form present in food, reflecting the potential health benefits associated
with the consumption of the entire fruit. At the same time, it is also important to identify
10
their metabolites and test their respective biological activities, since the hydrolysis of
glycosides and further gut bacterial transformation of aglycones may lead to the
production of more or less biologically active compounds, which may protect against
carcinogenesis (100, 119).
Apoptosis
Apoptosis or Programmed cell death is a physiological process that leads to
cellular self-destruction, which is an essential process in development, maintenance of
homeostasis and host defense in multicellular organisms. Apoptosis involves typical
morphological characteristics including plasma membrane blebbing, cell shrinkage,
nuclear chromatin condensation and fragmentation. In contrast to necrosis, apoptosis is
a tightly regulated process, which requires the activation of the intracellular machinery
by energy requiring (120). Dysregulation of this process contribute to various diseases,
including cancer (121-122).
A family of cystein-dependent aspartate-directed proteases, called caspases,
plays an essential role in apoptosis. Caspases are synthesized in normal cells as inactive
proenzymes (123), and can be activated by apoptosis signaling, which includes
autoproteolic cleavage or cleavage of other caspases at specific residues of aspartic acid
(124). During apoptosis, initiator caspases function as upstream signal transducers and
proteolytically activate downstream caspases (‘effector’ caspases). The initiator
caspases-2, -8, -9 and -10 are involved in interactions with adaptor proteins, whereas
effector caspases-3, -6 and -7 act on a variety of substrates resulting in proteolysis of
cellular proteins, leading to the apoptotic features (internucleosomal DNA
fragmentation, and culminating in cell death by apoptosis. Two pathways involved in
the activation of caspases have been extensivelly studied: the extrinsic death receptor
pathway, and the intrinsic or mitochondrial pathway (125-129).
Extrinsic death receptor pathway
Extrinsic death receptor pathway is characterized by external apoptosis
signalling, which is induced by several plasma membrane receptors belong to the
11
tumour necrosis factor (TNF)-receptor superfamily. This family includes Fas (Apo-1 or
CD95), TNF-receptor-1 (TNF-R1), death receptor-3, TNF-related apoptosis inducing
ligand receptor-1, TRAIL-R2 and DR6.
The death receptor Fas leads to receptor trimerisation and recruitment of specific
adaptor proteins to form an apoptotic signaling complex (130-131). The apoptosis
signaling mediated by Fas involves the ligation of a death ligand (Fas ligant) to the
transmembrane death receptor (Fas), which contains a death domain (DD) in cytoplasm
region that interacts with the adaptor protein (FADD), forming a death-receptor-induced
signalling complex (DISC). FADD, which contains a death effector domain (DED),
recruits the pro-caspase 8 into the disc, which will be proteolytically activated into
caspase-8. Thus, the effector caspase (Caspase-8) is an initiator of the caspase cascade,
because activate downstream effector caspases, including the apoptosis executioners
pro-caspases -3 and -7 into caspases -3 and -7, which cleavage proteins and activate
others caspases, resulting in cell death by apoptosis (129). Caspases -8 and -3 can
establish a link between the death receptor and the mitochondrial pathways by cleavage
and activation of Bid, which translocates from the cytosol to mitochondria and leads to
the release of cytochrome-c and apoptosis induction (132). The overview of Fasmediated extrinsic death receptor pathway is presented in Figure 3.
12
Fas ligant
Extrinsic death receptor pathway
Death receptor (Fas)
Cell membrane
FADD
Pro-caspases-8 or -10
Intrinsic pathway
DISC
Caspases-8 or -10
Mitochondrion
tBid
Pro-caspases-3 or -7
Pro-survival
Bcl-2 family
(eg. Bcl-2)
Pro-apoptotic
Bcl-2 family
(eg. Bax, Bim)
Caspase-3
Caspase-7
Bid
Cyrochrome c
Proteolysis
Apaf-1
Caspase-9
Cyrochrome c
Apaf-1
Pro-caspase-6
Caspase-6
Pro-caspase-9
Apaf-1
Cyrochrome c
Apoptosome
Figure 3. Pathways involved in apoptosis signaling: Fas-mediated extrinsic death receptor pathway and mitochondrial pathway; (
inhibition; (
) activation.
13
)
Mitochondrial pathway
The second pathway involves the participation of mitochondria in apoptosis,
which release caspase-activating proteins into the cytosol (Figure 3). The release of
cytochrome c from the mitochondria results in the activation of the apoptotic protease
activating factor-1 (Apaf-1). In the presence of cytochrome c and ATP, Apaf-1 bind and
activate procaspase-9, thus forming the mitochondrial DISC, also designated as
‘apoptosome’ (133). Following activation, the apoptosome-associated caspase-9 will in
turn activate downstream caspases like caspase- 3, -6 and -7 (132).
Intrinsic or mitochondria pathway is mainly regulated by proteins members of
the B cell lymphoma (Bcl-2) family, which plays multiple roles in carcinogenesis
process. These proteins normally reside in the intermembrane space of mitochondria
and, in response to a variety of apoptotic stimuli, can be released to the cytosol. It has
been suggested that changes in the concentration or function of Bcl-2 protein family
may lead to the cytochrome c release by modulating the permeabilization of the inner
and/or outer mitochondria membranes (134-135).
Bcl-2 family includes pro-apoptotic proteins, which facilitate this physiological
process of cell death, and pro-survival proteins, that inhibit apoptosis (136). The
homology between members of the Bcl-2 family is greatest within four small segments,
designated Bcl-2 homology (BH) regions, some of which contribute to the interactions
between Bcl-2 family members. Even though conclusion regarding whether the ability
to associate with other family members is central to apoptosis regulation, previous
studies suggested that the ability of Bcl-2 in inhibiting cell death requires binding to a
pro-apoptotic family member. Bcl-2 family can be divided into three groups according
to BH domains, mitochondrial anchorage (MA) and pro or anti-apoptotic action of their
proteins. They consist in anti-apoptotic (Bcl-2 and Bcl-xL) members, which contain
BH1, BH2, BH3 and BH4 domains; pro-apoptotic members with multidomains
including BH1, BH2 and BH3 (Bax and Bak), and citosolic BH3-only pro-apoptotic
members (Bid, Bad and Bim) (137-138).
Bax is known to induce apoptosis by interacting with pro-apoptotic members,
(Degli Esposti and Dive, 2003; Putcha et al., 2002). This protein forms channels and
contributes to regulate preexisting channels that permeabilize the outer mitochondrial
14
membrane, thus releasing caspase-activating proteins into the cytosol (139-140).
Likewise, Bim acts as sensor for cytoskeleton integrity and induce apoptosis by
neutralizing certain pro-survival Bcl-2 sub-family of proteins (141). Recently, it was
demonstrated the direct interaction between Bim and Bax domains, which suggest a
possible Bax activation by Bim (142).
Cell cycle regulation
Cell cycle consists in the series of events which occur in a cell that lead to its
division and replication. In eukaryotes, cell cycle is divided in interphase, process of
high expression of cell growth proteins which are required to mitosis; and mitosis, the
process of nuclear division. Mitosis includes four stages: prophase, metaphase,
anaphase and telophase, whereas interphase includes G1, S and G2 phases. DNA
Replication occurs in S phase, which is preceded by a gap called G1, a phase
characterized by cell growth and high biosynthetic activity of enzymes that are required
in S phase. Likewise, RNA transcription and protein synthesis rates are high in G2
phase, during which the cell prepares for mitosis (143-144). Cells in G1 can, before
commitment to DNA replication, enter a resting state called G0. The major part of the
non-growing, non-proliferating cells in the human body is found in G0 phase Although
the duration of cell cycle in tumor cells is equal to or longer than that of normal cell
cycle, the proportion of cells that are in active cell division (versus quiescent cells in G0
phase) in tumors is much higher than that in normal tissue. Thus there is a net increase
in cell number, whereas the number of cells that die by apoptosis or senescence may
remain the same (145).
Cell cycle, a highly conserved and regulated process, is essential to cell survival
because includes the repair of genetic damage as well as the prevention of uncontrolled
cell division. The mutation in some genes is the main cause of cell cycle dysregulation,
which may lead to an uncontrolled cellular proliferation, tumor formation and
carcinogenesis development (143, 146).
The transition from one cell cycle phase to another is regulated by cell cycle
genes and their protein products, which include cyclins, cyclin dependent kinases
(Cdks), Cdk inhibitors (CKI) and extracellular factors (growth factors) (147). The
15
regulation mechanisms involve controlled expression and destruction of cyclins,
activation and inhibitory phosphorylation and dephosphorylation of the cyclindependent kinases (Cdks), cyclins, and other proteins; the binding of a number of Cdk
inhibitory proteins; and the expression and destruction of inhibitory proteins associated
with Cdks, or Cdk/cyclin complexes (143, 146, 148-150).
Cyclins are targets for extracellular signaling and frequently are dysregulated
during oncogenesis. These proteins are produced or degraded as needed during
interphase in order to drive the cell through the different stages of cell cycle. They
control the progression of cell cycle by activating cyclin-dependent kinase (Cdk)
enzymes. Cyclins consist in cell cycle regulators which are activated by Cdks at specific
points: at the G1 phase (Cyclin D), at G1/S transition (Cyclin E), at S phase (Cyclin A),
at G2/M phase transition ( Cyclin A) and in mitosis (Cyclin B) (145, 151).
Likewise, cyclin-dependent kinases are cell cycle regulatory proteins from a
serine/threonine protein kinases family. Some of these proteins are known to induce
downstream processes by phosphorylating specific proteins upon becoming active by
forming clyclin complexes with other proteins, or by phosphorylation during different
cell cycle phases (148). In humans, Cdk4, Cdk6, Cdk2 are know to regulate G1 phase,
Cdk2 is active at S phase, while Cdk1 controls mitosis (152).
Phosphorylation plays a central role in regulating the formation, activation and
inactivation of CDK/cyclin complexes. Protein kinases phosphorylate proteins involved
in mRNA production, such as cyclin-dependent Kinases (CDKs), which become active
upon binding to a cyclin (153). In mammalians, cell division control protein 2 homolog
(p34, cdc2, cyclin B or Cdk1) is a cyclin-dependend kinase from the serine/threonine
protein kinase family, which is encoded by the CDC2 gene (154). This protein is a
catalytic subunit of the highly conserved protein kinase complex known as maturation
promoting factor (MPF), composed of cyclin B and Cdk1 (cdc2). MPF controls the
transition from G2/M phase to mitosis by phosphorylating multiple proteins needed
during mitosis. Thus, these phosphorylated proteins are responsible for specific events
during cycle division such as microtubule formation and chromatin remodeling (155156). When complexed to cyclins, Cdc2 can be phosphorilated and inactivated by the
membrane-associated
tyrosine-
and
threonine-specific
cdc2-inhibitory
kinase
(PKMYT1), an enzyme of the serine/threonine protein kinase family which is encoded
16
by the PKMYT1 gene in humans (157). Therefore, this enzyme plays an important role
in carcinogenesis, because negatively regulates cell cycle G2/M transition by cdc-2
inactivation (157), which may leads to G2/M cell cycle arrest and cell growth
suppression.
The activity of cyclin-dependent kinase (Cdks) can be counteracted by cell cycle
inhibitory proteins, known as cyclin-dependent kinase inhibitors (CKI), which are
produced by genes called tumor suppressor genes. CKIs can halt cell cycle and,
consequently, the tumor progression, by binding Cdk alone or Cdk/cyclin complexes.
CKIs are distributed in two major families: the INK4a/ARF (Inhibitor of Kinase
4/Alternative Reading Frame) family, composed of inhibitors (p15, p16, p18 and p19)
of single Cdks enzyme before cyclin binding (158); and the cip/kip family, composed of
CDKs (p21, p27 and p57) that inactivate CDK-cyclin complexes (159). In addition, they
inhibit the CDK/cyclin complexes at G1 phase, and to a lesser extent, Cdk1/cyclin B
complexes (160).
The cyclin-dependent kinase inhibitor 1A (Cip1/WAF1 or p21), a protein
encoded by CDKN1A gene, mainly regulate cell cycle at G1 phase by inhibiting the
activity of Cyclin/Cdk2 complexes (161). The expression of p21 is mainly under
transcriptional control of the p53 tumor suppressor gene, although may be activated by a
response to a variety of stress stimuli (162). When overexpressed, p21 showed to induce
cell cycle arrest at G1 (161), G2 (163) and S (164) phases of cancer cells, which
contributed to cell growth suppression (165-167). In addition to cell cycle arrest, p21
expression can mediate cellular senescence (G0 phase) by interacting with proliferating
cell nuclear antigen (PCNA), a DNA polymerase accessory factor, thus blocking DNA
synthesis during cell cycle progression at S phase (168). Additionally, this protein was
reported to induce apoptosis because p21 can be specifically cleaved by caspase-3,
which thus leads to a dramatic activation of CDK2 and caspase cascade activation
(166). On the other hand, when p21 is repressed, different results can appear depending
on the context. Studies have shown that p21 repression contributed or not to cancer
progression by apoptosis inhibition. However, the mechanism involved is still not
completely understood (165).
17
Anticarcinogenic effects of polyphenols in normal as compared to cancer
cells
Tumor cells present several aspects which make them different from normal
cells. Cancerous cells do not depend on growth factors like normal cells do, because
they are able to produce their own growth factors to stimulate cell proliferation. Normal
cells need to keep in contact with the extracellular medium to growth, whereas cancer
cells are not dependent on anchorage. Moreover, in cell culture, normal cells growth in
a monolayer and may be inhibited by closed cells, whereas cancerous cells growth to
form several monolayers. Tumoral cells present less adherence than normal cells.
Additionally, normal cells interrupt cell proliferation as soon as they reach certain
density, whereas tumoral cells keep growing. Normal cells remain in the area where
they belong and do not spread to other parts of the body. Cancer cells, otherwise, may
spread through the body (metastasize), by direct invasion and destruction of the organ of
origin, or spread through the lymphatic system or bloodstream to distant organs such as
the bone, lung, and liver (10).
Several in vitro studies have been demonstrated the chemopreventive effects of
polyphenols in cancer cells but not in normal cells (169-173). Previous studies have
reported that growth-inhibitory effects of polyphenols were greater in colon cancer as
compared to non-tumorigenic colon cells (169-170). The isogenic human colon cancer
cell lines HCT-116(p53 +/+) (IC50 value
value
45 μg/mL) and HCT-116 (p53 -/-) (IC50
30 μg/mL) showed to be more sensitive to the treatment with gallotannins
than normal human intestinal epithelial cells FHs 74Int (IC50 value > 60 μg/mL) (173).
Overall, cancer cells have been demonstrated to be more sensitive to polyphenolinduced apoptosis than normal cells, which often undergo apoptosis only with higher
polyphenol-concentrations. Although the mechanism for this phenomenon are still not
well elucidated, possible differences between normal and cancer cells may explain in
part the higher sensibility of cancer cells. These include membrane fluidity, expression
and activity of non-mutated proteins, oncogenes, as well as intracellular signal
transduction (174-177).
18
Objectives
The overall objective of this study was to elucidate the anti-cancer effects of
mango polyphenols of several varieties in different types of cancer.
Based on previous published data which state that individual polyphenols found in
mango pulp present anticancer properties including antiproliferative effects, induction
of apoptosis, regulation of cell cycle and inhibition of angiogenesis, we hypothesize that
mango phenolic extracts present chemopreventive properties, and might represent the
health benefits derived from the consumption of the entire fruit instead of isolated
compounds. The antiproliferative effects in vitro of polyphenols from different mango
varieties (Francis, Kent, Ataulfo, Tommy Atkins and Haden) were compared on
different cancer cell lines (Molt-4 leukemia, A-549 lung, MDA-MB-231 breast, LnCap
prostate, SW-480 colon cancer cells and the non-cancer cell line CCD-18Co).
Molecular mechanisms involved on the anti-cancer activities of mango polyphenols on
colon cancer cells were assessed by studying the effect of mango polyphenols on gene
expression, cell cycle regulation and reactive oxygen species production.
Based on previous publications indicating the increased bioavailability of
polyphenolics after metabolism by β-glucosidase, we hypothesize that the enzymatic
hydrolysis of mango polyphenols could enhance their absorption in a selected cell
culture model. This hypothesis was tested using Caco-2 human coloncarcinoma cell
monolayers in vitro model. In order to elucidate the in vivo intestinal absorption, mango
polyphenols were prior hydrolyzed using the enzyme β-glucosidase, commonly found in
gut microflora. The effect of enzymatic hydrolysis on antioxidant activity, phenolic
content and cell-growth suppressive activity of mango polyphenols was evaluated on
MDA-MB-231 breast and HT-29 colon human cancer cells. The antiproliferative
activity of high molecular weight polyphenols-rich fraction, characterized by mangiferin
(422.35 Da) and gallotannins (788-1851 Da), and low molecular weight polyphenolsrich fraction, comprising mainly phenolic acids (138-194 Da), was studied using cell
culture models.
Results from this study provide valuable insight into the anti-cancer effects of
mango polyphenols and the mechanisms underlying those effects. Furthermore,
information regarding the antioxidant and cancer-inhibitory activities of mango
19
polyphenols contributes to the ongoing evaluation of health benefits derived from fruit
consumption.
Overall, the performed studies elucidate the preventive effects of polyphenols
extracted from mango on carcinogenesis process.
20
REFERENCES
1.
WHO, World Health Statistics 2008, World Health Organization: Geneva
Switzerland, 2008.
2.
American
Cancer
Society
Cancer
Facts
&
Figures
2008
http://www.cancer.org/downloads/STT/2008CAFFfinalsecured.pdf (03/13/09).
3.
INCA, Estimativas 2010: Incidência de Câncer no Brasil. 2009, National
Institutto of Cancer http://www1.inca.gov.br/vigilancia/incidencia.html (28-11-09).
4.
Brasilerio Fillho, G., Patologia Geral 3. ed. Rio de Janeiro: Guanabara Koogan,
2004.
5.
Harnden, D. G.; Mcgee, J. O. D., Textbook of Patology 1992, Oxford University
Press v.1, 571-717.
6.
Soussi, T., Cancer epidemiology: from the description to molecular biology. M
S-Med Sci 2000, 16, (12), 1397-1404.
7.
Bogliolo, L.; Pereira, F. E. L.; Guimaraes, R. C., Bogliolo Patologia 1987, 4. ed.
Rio de Janeiro: Guanabara Koogan, 180-205.
8.
Cotran, R. S.; Kumar, V.; Collins, T. R., Patologia Estrutural e Funcional.
2000, 6. ed. Rio de Janeiro: Guanabara Koogan, 233-293.
9.
Coussens, L. M.; Werb, Z., Inflammation and cancer. Nature 2002, 20, 860-867.
10.
Hanahan, D.; Weinberg, R. A., The hallmarks of cancer. Cell 2000, 100, (1), 57-
70.
11.
Shay, J. W.; Roninson, I. B., Hallmarks of senescence in carcinogenesis and
cancer therapy. Oncogene 2004, 23, (16), 2919-2933.
12.
Vattemi, E.; Claudio, P. P., Tumor suppressor genes as cancer therapeutics.
Drug News Perspect 2007, 20, (8), 511-520.
13.
Burzynski, S. R., Aging: gene silencing or gene activation? Med Hypotheses
2005, 64, (1), 201-208.
14.
Ross, D. W., Cancer: The emerging molecular biology. Hosp Pract 2000, 35,
(1), 63-+.
15.
Mertens-Talcott, S. U.; Bomser, J. A.; Romero, C.; Talcott, S. T.; Percival, S. S.,
Ellagic acid potentiates the effect of quercetin on p21(waf1/cip1), p53, and MAP-
21
kinases without affecting intracellular generation of reactive oxygen species in vitro. J
Nutr 2005, 135, (3), 609-614.
16.
Mertens-Talcott, S. U.; Bomser, J. A.; Talcott, S. T.; Percival, S. S., Quercetin
and ellagic acid act synergistically in the induction of apoptosis and p21(WAF1/C1P1)involved signal transduction in human MOLT-4 leukemia cells. Faseb J 2004, 18, (4),
A379-A379.
17.
Mertens-Talcott, S. U.; Chintharlapalli, S.; Li, M. R.; Safe, S., The oncogenic
microRNA-27a targets genes that regulate specificity protein transcription factors and
the G(2)-M checkpoint in MDA-MB-231 breast cancer cells. Cancer Res 2007, 67, (22),
11001-11011.
18.
Mertens-Talcott, S. U.; Jilma-Stohlawetz, P.; Rios, J.; Hingorani, L.; Derendorf,
H., Absorption, metabolism, and antioxidant effects of pomegranate (Punica granatum
L.) polyphenols after ingestion of a standardized extract in healthy human volunteers. J
Agr Food Chem 2006, 54, (23), 8956-8961.
19.
De Castro, W. V.; Mertens-Talcott, S.; Derendorf, H.; Butterweck, V.,
Grapefruit juice-drug interactions: Grapefruit juice and its components inhibit pglycoprotein (ABCB1) mediated transport of talinolol in caco-2 cells. J Pharm Sci-Us
2007, 96, (10), 2808-2817.
20.
Ross, D. T.; Scherf, U.; Eisen, M. B.; Perou, C. M.; Rees, C.; Spellman, P.; Iyer,
V.; Jeffrey, S. S.; Van de Rijn, M.; Waltham, M.; Pergamenschikov, A.; Lee, J. C. E.;
Lashkari, D.; Shalon, D.; Myers, T. G.; Weinstein, J. N.; Botstein, D.; Brown, P. O.,
Systematic variation in gene expression patterns in human cancer cell lines. Nat Genet
2000, 24, (3), 227-235.
21.
Block, K. I.; Koch, A. C.; Mead, M. N.; Tothy, P. K.; Newman, R. A.;
Gyllenhaal, C., Impact of antioxidant supplementation on chemotherapeutic efficacy: A
systematic review of the evidence from randomized controlled trials. Cancer Treat Rev
2007, 33, (5), 407-418.
22.
Abdulla, M.; Gruber, P., Role of diet modification in cancer prevention.
Biofactors 2000, 12, (1-4), 45-51.
23.
Bialy, T. L.; Rothe, M. J.; Grant-Kels, J. M., Dietary factors in the prevention
and treatment of nonmelanoma skin cancer and melanoma. Dermatologic Surgery 2002,
28, (12), 1143-1152.
22
24.
Moure, A.; Cruz, J. M.; Franco, D.; Dominguez, J. M.; Sineiro, J.; Dominguez,
H.; Nunez, M. J.; Parajo, J. C., Natural antioxidants from residual sources. Food Chem
2001, 72, (2), 145-171.
25.
Shahidi, F.; Naczk, M., Food phenolics: souces, chemistry, effects and
applications. 1. ed. Lancaster: Technomic Publishing Co, Inc. 1995, 331p.
26.
Scalbert, A.; Williamson, G., Dietary intake and bioavailability of polyphenols.
J Nutr 2000, 130, (8), 2073s-2085s.
27.
Amin, A. R. M. R.; Kucuk, O.; Khuri, F. R.; Shin, D. M., Perspectives for
Cancer Prevention With Natural Compounds. J Clin Oncol 2009, 27, (16), 2712-2725.
28.
Korkina, L. G.; De Luca, C.; Kostyuk, V. A.; Pastore, S., Plant Polyphenols and
Tumors: From Mechanisms to Therapies, Prevention, and Protection Against Toxicity
of Anti-Cancer Treatments. Curr Med Chem 2009, 16, (30), 3943-3965.
29.
Brisdelli, F.; D'Andrea, G.; Bozzi, A., Resveratrol: A Natural Polyphenol with
Multiple Chemopreventive Properties (Review). Curr Drug Metab 2009, 10, (6), 530546.
30.
Magrone, T.; Candore, G.; Caruso, C.; Jirillo, E.; Covelli, V., Polyphenols from
Red Wine Modulate Immune Responsiveness: Biological and Clinical Significance.
Curr Pharm Design 2008, 14, (26), 2733-2748.
31.
Selma, M. V.; Espin, J. C.; Tomas-Barberan, F. A., Interaction between
Phenolics and Gut Microbiota: Role in Human Health. J Agr Food Chem 2009, 57, (15),
6485-6501.
32.
Parvez, S.; Tabassum, H.; Rehman, H.; Banerjee, B. D.; Athar, M.; Raisuddin,
S., Catechin prevents tamoxifen-induced oxidative stress and biochemical perturbations
in mice. Toxicology 2006, 225, (2-3), 109-118.
33.
Yamamoto, M.; Miyamoto, S.; Moon, J. H.; Murota, K.; Hara, Y.; Terao, J.,
Effect of dietary green tea catechin preparation on oxidative stress parameters in large
intestinal mucosa of rats. Biosci Biotech Bioch 2006, 70, (1), 286-289.
34.
Huang, Q.; Wu, L. J.; Tashiro, S.; Onodera, S.; Ikejima, T., Elevated levels of
DNA repair enzymes and antioxidative enzymes by (+)-catechin in murine microglia
cells after oxidative stress. J Asian Nat Prod Res 2006, 8, (1-2), 61-71.
23
35.
Mertens-Talcott, S. U.; Talcott, S. T.; Percival, S. S., Low concentrations of
quercetin and ellagic acid synergistically influence proliferation, cytotoxicity and
apoptosis in MOLT-4 human leukemia cells. J Nutr 2003, 133, (8), 2669-2674.
36.
Mertens-Talcott, S. U.; Percival, S. S., Ellagic acid and quercetin interact
synergistically with resveratrol in the induction of apoptosis and cause transient cell
cycle arrest in human leukemia cells. Cancer Lett 2005, 218, (2), 141-151.
37.
FAOSTAT, FAO Statistical Database – Agriculture. http://apps.fao.org ( 11-09-
2009) 2009.
38.
Masibo, M.; He, Q., Major mango polyphenols and their potential significance
to human health. Compr Rev Food Sci F 2008, 7, (4), 309-319.
39.
Camargo Filho, W. P. d.; Alves, H. S.; Mazzei, A. R., Mercado de manga no
Brasil: contexto mundial, variedades e estacionalidade Informações Econômicas, SP
<http://www.iea.sp.gov.br/OUT/publicacoes/pdf/tec4-0504.pdf> (11-11-2009). 2004,
34, (5).
40.
Garrido, G.; Gonzalez, D.; Lemus, Y.; Garcia, D.; Lodeiro, L.; Quintero, G.;
Delporte, C.; Nunez-Selles, A. J.; Delgado, R., In vivo and in vitro anti-inflammatory
activity of Mangifera indica L. extract (VIMANG (R)). Pharmacol Res 2004, 50, (2),
143-149.
41.
Garrido, G.; Delgado, R.; Lemus, Y.; Rodriguez, J.; Garcia, D.; Nunez-Selles, A.
J., Protection against septic shock and suppression of tumor necrosis factor alpha and
nitric oxide production on macrophages and microglia by a standard aqueous extract of
Mangifera indica L. (VIMANG (R)) - Role of mangiferin isolated from the extract.
Pharmacol Res 2004, 50, (2), 165-172.
42.
Garrido, G.; Gonzalez, D.; Delporte, C.; Backhouse, N.; Quintero, G.; Nunez-
Selles, A. J.; Morales, M. A., Analgesic and anti-inflammatory effects of Mangifera
indica L. extract (Vimang). Phytother Res 2001, 15, (1), 18-21.
43.
Martinez, G.; Delgado, R.; Perez, G.; Garrido, G.; Selles, A. J. N.; Leon, O. S.,
Evaluation of the in vitro antioxidant activity of Mangifera indica L. extract (Vimang).
Phytother Res 2000, 14, (6), 424-427.
44.
Nunez-Selles, A. J.; Delgado-Hernandez, R.; Garrido-Garrido, G.; Garcia-
Rivera, D.; Guevara-Garcia, M.; Pardo-Andreu, G. L., The paradox of natural products
24
as pharmaceuticals - Experimental evidences of a mango stem bark extract. Pharmacol
Res 2007, 55, (5), 351-358.
45.
Nunez-Selles, A. J., Antioxidant therapy: Myth or reality? J Brazil Chem Soc
2005, 16, (4), 699-710.
46.
Schieber, A.; Ullrich, W.; Carle, R., Characterization of polyphenols in mango
puree concentrate by HPLC with diode array and mass spectrometric detection.
Innovative Food Science and Emerging Technologies 2000, 1, (2), 161-166.
47.
Berardini, N.; Carle, R.; Schieber, A., Characterization of gallotannins and
benzophenone derivatives from mango (Mangifera indica L. cv. 'Tommy Atkins') peels,
pulp and kernels by high-performance liquid chromatography electrospray ionization
mass spectrometry. Rapid Commun Mass Sp 2004, 18, (19), 2208-2216.
48.
Barreto, J. C.; Trevisan, M. T. S.; Hull, W. E.; Erben, G.; de Brito, E. S.;
Pfundstein, B.; Wurtele, G.; Spiegelhalder, B.; Owen, R. W., Characterization and
quantitation of polyphenolic compounds in bark, kernel, leaves, and peel of mango
(Mangifera indica L.). J Agr Food Chem 2008, 56, (14), 5599-5610.
49.
Huh, J. E.; Lee, E. O.; Kim, M. S.; Kang, K. S.; Kim, C. H.; Cha, B. C.; Surh, Y.
J.; Kim, S. H., Penta-O-galloyl-beta-D-glucose suppresses tumor growth via inhibition
of angiogenesis and stimulation of apoptosis: Roles of cyclooxygenase-2 and mitogenactivated protein kinase pathways. J Vasc Res 2006, 43, 75-75.
50.
Zhang, J. H.; Li, L.; Kim, S. H.; Hagerman, A. E.; Lu, J. X., Anti-Cancer, Anti-
Diabetic and Other Pharmacologic and Biological Activities of Penta-Galloyl-Glucose.
Pharm Res-Dord 2009, 26, (9), 2066-2080.
51.
Chen, W. J.; Chang, C. Y.; Lin, J. K., Induction of G1 phase arrest in MCF
human breast cancer cells by pentagalloylglucose through the down-regulation of CDK4
and CDK2 activities and up-regulation of the CDK inhibitors p27(Kip) and p21(Cip).
Biochem Pharmacol 2003, 65, (11), 1777-1785.
52.
Chen, W. J.; Lin, J. K., Induction of G1 arrest and apoptosis in human jurkat T
cells by pentagalloylglucose through inhibiting proteasome activity and elevating
p27Kip1, p21Cip1/WAF1, and Bax proteins. . J Biol Chem 2004 2004, 279, (14), 496505.
53.
Oh, G. S.; Pae, H. O.; Oh, H.; Hong, S. G.; Kim, I. K.; Chai, K. Y.; Yun, Y. G.;
Kwon, T. O.; Chung, H. T., In vitro anti-proliferative effect of 1,2,3,4,6-penta-O-
25
galloyl-beta-D-glucose on human hepatocellular carcinoma cell line, SK-HEP-1 cells.
Cancer Lett 2001, 174, (1), 17-24.
54.
Liu, Z. J.; Schwimer, J.; Liu, D.; Lewis, J.; Greenway, F. L.; York, D. A.;
Woltering, E. A., Gallic acid is partially responsible for the antiangiogenic activities of
Rubus leaf extract. Phytother Res 2006, 20, (9), 806-813.
55.
Jagetia, G. C.; Venkatesha, V. A., Effect of mangiferin on radiation-induced
micronucleus formation in cultured human peripheral blood lymphocytes. Environ Mol
Mutagen 2005, 46, (1), 12-21.
56.
Jagetia, G. C.; Baliga, M. S., Radioprotection by mangiferin in DBAxC(57)BL
mice: a preliminary study. Phytomedicine 2005, 12, (3), 209-215.
57.
Leiro, J. M.; Alvarez, E.; Arranz, J. A.; Siso, I. G.; Orallo, F., In vitro effects of
mangiferin on superoxide concentrations and expression of the inducible nitric oxide
synthase, tumour necrosis factor-alpha and transforming growth factor-beta genes.
Biochem Pharmacol 2003, 65, (8), 1361-1371.
58.
Yoshimi, N.; Matsunaga, K.; Katayama, M.; Yamada, Y.; Kuno, T.; Qiao, Z.;
Hara, A.; Yamahara, J.; Mori, H., The inhibitory effects of mangiferin, a naturally
occurring glucosylxanthone, in bowel carcinogenesis of male F344 rats. Cancer Lett
2001, 163, (2), 163-170.
59.
Rajendran, P.; Ekambaram, G.; Sakthisekaran, D., Effect of mangiferin on
benzo(a)pyrene induced lung carcinogenesis in experimental Swiss albino mice. Nat
Prod Res 2008, 22, (8), 672-680.
60.
Pardo-Andreu, G. L.; Dorta, D. J.; Delgado, R.; Cavalheiro, R. A.; Santos, A. C.;
Vercesi, A. E.; Curti, C., Vimang (Mangifera indica L. extract) induces permeability
transition in isolated mitochondria, closely reproducing the effect of mangiferin,
Vimang's main component. Chem-Biol Interact 2006, 159, (2), 141-148.
61.
Chaturvedi, P. K.; Bhui, K.; Shukla, Y., Lupeol: Connotations for
chemoprevention. Cancer Lett 2008, 263, (1), 1-13.
62.
Faried, A.; Kurnia, D.; Faried, L. S.; Usman, N.; Miyazaki, T.; Kato, H.;
Kuwano, H., Anticancer effects of gallic acid isolated from Indonesian herbal medicine,
Phaleria macrocarpa (Scheff.) Boerl, on human cancer cell lines. Int J Oncol 2007, 30,
(3), 605-613.
26
63.
Serrano, A.; Palacios, C.; Roy, G.; Cespon, C.; Villar, M. L.; Nocito, M.;
Gonzalez-Porque, P., Derivatives of gallic acid induce apoptosis in tumoral cell lines
and inhibit lymphocyte proliferation. Arch Biochem Biophys 1998, 350, (1), 49-54.
64.
Salucci, M.; Stivala, L. A.; Maiani, G.; Bugianesi, R.; Vannini, V., Flavonoids
uptake and their effect on cell cycle of human colon adenocarcinoma cells (Caco2). Brit
J Cancer 2002, 86, (10), 1645-1651.
65.
Huh, J. E.; Lee, E. O.; Kim, M. S.; Kang, K. S.; Kim, C. H.; Cha, B. C.; Surh, Y.
J.; Kim, S. H., Penta-O-galloyl-beta-D-glucose suppresses tumor growth via inhibition
of angiogenesis and stimulation of apoptosis: roles of cyclooxygenase-2 and mitogenactivated protein kinase pathways. Carcinogenesis 2005, 26, (8), 1436-1445.
66.
Percival, S. S.; Talcott, S. T.; Chin, S. T.; Mallak, A. C.; Lounds-Singleton, A.;
Pettit-Moore, J., Neoplastic transformation of BALB/3T3 cells and cell cycle of HL-60
cells are inhibited by mango (Mangifera indica L.) juice and mango juice extracts. J
Nutr 2006, 136, (5), 1300-1304.
67.
Garcia-Solis, P.; Yahia, E. M.; Morales-Tlalpan, V.; Diaz-Munoz, M., Screening
of antiproliferative effect of aqueous extracts of plant foods consumed in Mexico on the
breast cancer cell line MCF-7. . Int J Food Sci Nutr 2009, 1-15.
68.
Garcia-Solis, P.; Yahia, E. M.; Aceves, C., Study of the effect of 'Ataulfo'
mango (Mangifera indica L.) intake on mammary carcinogenesis and antioxidant
capacity in plasma of N-methyl-N-nitrosourea (MNU)-treated rats. Food Chem 2008,
111, (2), 309-315.
69.
Prasad, S.; Kalra, N.; Shukla, Y., Induction of apoptosis by lupeol and mango
extract in mouse prostate and LNCaP cells. Nutr Cancer 2008, 60, (1), 120-130.
70.
Prasad, S.; Kalra, N.; Singh, M.; Shukla, Y., Protective effects of lupeol and
mango extract against androgen induced oxidative stress in Swiss albino mice. Asian J
Androl 2008, 10, (2), 313-318.
71.
Ribeiro, S. M. R., Caracterização e avaliação do potencial antioxidante de
mangas (Mangifera indica L.) cultivadas no Estado de Minas Gerais. PhD Dissertation –
Universidade Federal de Viçosa, Viçosa, M.G. 2006.
72.
Balsano, C.; Alisi, A., Antioxidant Effects of Natural Bioactive Compounds.
Curr Pharm Design 2009, 15, (26), 3063-3073.
27
73.
Surh, Y. J., Cancer chemoprevention with dietary phytochemicals. Nat Rev
Cancer 2003, 3, (10), 768-780.
74.
Auten, R. L.; Davis, J. M., Oxygen Toxicity and Reactive Oxygen Species: The
Devil Is in the Details. Pediatric Research 2009, 66, (2), 121-127.
75.
Afonso, V.; Champy, R.; Mitrovic, D.; Collin, P.; Lomri, A., Reactive oxygen
species and superoxide dismutases: Role in joint diseases. Joint Bone Spine 2007, 74,
(4), 324-329.
76.
Martinez, M. C.; Andriantsitohaina, R., Reactive Nitrogen Species: Molecular
Mechanisms and Potential Significance in Health and Disease. Antioxid Redox Sign
2009, 11, (3), 669-702.
77.
Datta, K.; Sinha, S.; Chattopadhyay, P., Reactive oxygen species in health and
disease. National Medical Journal of India 2000, 13, (6), 304-310.
78.
Weinberg, F.; Chandel, N. S., Reactive oxygen species-dependent signaling
regulates cancer. Cell Mol Life Sci 2009, 66, (23), 3663-3673.
79.
Behrend, L.; Henderson, G.; Zwacka, R. M., Reactive oxygen species in
oncogenic transformation. Biochem Soc T 2003, 31, 1441-1444.
80.
Feig, D. I.; Reid, T. M.; Loeb, L. A., Reactive Oxygen Species in
Tumorigenesis. Cancer Res 1994, 54, (7), S1890-S1894.
81.
Storz, P., Reactive oxygen species in tumor progression. Front Biosci 2005, 10,
1881-1896.
82.
Raza, H.; John, A., In Vitro Effects of Tea Polyphenols on Redox Metabolism,
Oxidative Stress, and Apoptosis in PC12 Cells. Recent Advances in Clinical Oncology
2008, 1138, 358-365.
83.
Hong, J.; Kim, M. R.; Lee, N. H.; Lee, B. H., Inhibition of Oral Epithelial Cell
Growth in vitro by Epigallocatechin-3-gallate; Its Modulation by Serum and
Antioxidant Enzymes. Food Sci Biotechnol 2009, 18, (4), 971-977.
84.
Hadi, S. M.; Bhat, S. H.; Azmi, A. S.; Hanif, S.; Shamim, U.; Ullah, M. F.,
Oxidative breakage of cellular DNA by plant polyphenols: A putative mechanism for
anticancer properties. Semin Cancer Biol 2007, 17, (5), 370-376.
85.
Azad, N.; Rojanasakul, Y.; Vallyathan, V., Inflammation and lung cancer: Roles
of reactive oxygen/nitrogen species. J Toxicol Env Heal B 2008, 11, (1), 1-15.
28
86.
Antosiewicz, J.; Ziolkowski, W.; Kar, S.; Powolny, A. A.; Singh, S. V., Role of
Reactive
Oxygen
Intermediates
in
Cellular
Responses
to
Dietary
Cancer
Chemoprevention Agents. Planta Med 2008, 74, (13), 1570-1579.
87.
Engel, R. H.; Evens, A. M., Oxidative stress and apoptosis: a new treatment
paradigm in cancer. Front Biosci 2006, 11, 300-312.
88.
Manach, C.; Scalbert, A.; Morand, C.; Remesy, C.; Jimenez, L., Polyphenols:
food sources and bioavailability. Am J Clin Nutr 2004, 79, (5), 727-747.
89.
Scalbert, A.; Manach, C.; Morand, C.; Remesy, C., Bioavailability of dietary
polyphenols is a key issue to assess their impact on physiological functions. Free
Radical Bio Med 2004, 36, S27-S27.
90.
Scalbert, A.; Morand, C.; Manach, C.; Remesy, C., Absorption and metabolism
of polyphenols in the gut and impact on health. Biomed Pharmacother 2002, 56, (6),
276-282.
91.
Nemeth, K.; Plumb, G. W.; Berrin, J. G.; Juge, N.; Jacob, R.; Naim, H. Y.;
Williamson, G.; Swallow, D. M.; Kroon, P. A., Deglycosylation by small intestinal
epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of
dietary flavonoid glycosides in humans. Eur J Nutr 2003, 42, (1), 29-42.
92.
Lee, H. C.; Jenner, A. M.; Low, C. S.; Lee, Y. K., Effect of tea phenolics and
their aromatic fecal bacterial metabolites on intestinal microbiota. Res Microbiol 2006,
157, (9), 876-884.
93.
Davis, C. D.; Milner, J. A., Gastrointestinal microflora, food components and
colon cancer prevention. J Nutr Biochem 2009, 20, (10), 743-752.
94.
Rechner, A. R.; Smith, M. A.; Kuhnle, G.; Gibson, G. R.; Debnam, E. S.; Srai,
S. K. S.; Moore, K. P.; Rice-Evans, C. A., Colonic metabolism of dietary polyphenols:
Influence of structure on microbial fermentation products. Free Radical Bio Med 2004,
36, (2), 212-225.
95.
Lampe, J. W.; Chang, J. L., Interindividual differences in phytochemical
metabolism and disposition. Semin Cancer Biol 2007, 17, (5), 347-353.
96.
Clark, D. E., Rapid calculation of polar molecular surface area and its
application to the prediction of transport phenomena. 1. Prediction of intestinal
absorption. J Pharm Sci-Us 1999, 88, (8), 807-814.
29
97.
Lipinski, C. A., Drug-like properties and the causes of poor solubility and poor
permeability. J Pharmacol Toxicol 2000, 44, (1), 235-249.
98.
Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J., Experimental and
computational approaches to estimate solubility and permeability in drug discovery and
development settings. Adv Drug Deliver Rev 2001, 46, (1-3), 3-26.
99.
Lee, C. Y.; Cheng, H. M.; Sim, S. M., Bioavailability of dietary flavonoids and
carotenoids. Curr Top Nutraceut R 2006, 4, (1), 33-51.
100.
Yang, C. S.; Landau, J. M.; Huang, M. T.; Newmark, H. L., Inhibition of
carcinogenesis by dietary polyphenolic compounds. Annu Rev Nutr 2001, 21, 381-406.
101.
Warden, B. A.; Smith, L. S.; Beecher, G. R.; Balentine, D. A.; Clevidence, B.
A., Catechins are bioavailable in men and women drinking black tea throughout the day.
J Nutr 2001, 131, (6), 1731-1737.
102.
Santos-Buelga, C.; Scalbert, A., Proanthocyanidins and tannin-like compounds -
nature, occurrence, dietary intake and effects on nutrition and health. J Sci Food Agr
2000, 80, (7), 1094-1117.
103.
Deprez, S.; Mila, I.; Huneau, J. F.; Tome, D.; Scalbert, A., Transport of
proanthocyanidin dimer, trimer, and polymer across monolayers of human intestinal
epithelial Caco-2 cells. Antioxid Redox Sign 2001, 3, (6), 957-967.
104.
Holt, R. R.; Lazarus, S. A.; Sullards, M. C.; Zhu, Q. Y.; Schramm, D. D.;
Hammerstone, J. F.; Fraga, C. G.; Schmitz, H. H.; Keen, C. L., Procyanidin dimer B2
[epicatechin-(4 beta-8)-epicatechin] in human plasma after the consumption of a
flavanol-rich cocoa. Am J Clin Nutr 2002, 76, (4), 798-804.
105.
Prasain, J. K.; Peng, N.; Dai, Y. Y.; Moore, R.; Arabshahi, A.; Wilson, L.;
Barnes, S.; Wyss, J. M.; Kim, H.; Watts, R. L., Liquid chromatography tandem mass
spectrometry identification of proanthocyanidins in rat plasma after oral administration
of grape seed extract. Phytomedicine 2009, 16, (2-3), 233-243.
106.
Appeldoorn, M. M.; Vincken, J. P.; Gruppen, H.; Hollman, P. C. H., Procyanidin
Dimers A1, A2, and B2 Are Absorbed without Conjugation or Methylation from the
Small Intestine of Rats. J Nutr 2009, 139, (8), 1469-1473.
107.
Appeldoorn, M. M.; Vincken, J. P.; Aura, A. M.; Hollman, P. C. H.; Gruppen,
H., Procyanidin Dimers Are Metabolized by Human Microbiota with 2-(3,4-
30
Dihydroxyphenyl)acetic Acid and 5-(3,4-Dihydroxyphenyl)-gamma-valerolactone as
the Major Metabolites. J Agr Food Chem 2009, 57, (3), 1084-1092.
108.
Cerda, B.; Llorach, R.; Ceron, J. J.; Espin, J. C.; Tomas-Barberan, F. A.,
Evaluation of the bioavailability and metabolism in the rat of punicalagin, an
antioxidant polyphenol from pomegranate juice. Eur J Nutr 2003, 42, (1), 18-28.
109.
Larrosa, M.; Tomas-Barberan, F. A.; Espin, J. C., The dietary hydrolysable
tannin punicalagin releases ellagic acid that induces apoptosis in human colon
adenocarcinoma Caco-2 cells by using the mitochondrial pathway. J Nutr Biochem
2006, 17, (9), 611-625.
110.
Konishi, Y.; Kobayashi, S.; Shimizu, M., Transepithelial transport of p-coumaric
acid and gallic acid in caco-2 cell monolayers. Biosci Biotech Bioch 2003, 67, (11),
2317-2324.
111.
Vaidyanathan, J. B.; Walle, T., Transport and metabolism of the tea flavonoid (-
)-epicatechin by the human intestinal cell line Caco-2. Pharm Res-Dord 2001, 18, (10),
1420-1425.
112.
Arafa, H. M. M., Possible contribution of beta-glucosidase and caspases in the
cytotoxicity of glufosfamide in colon cancer cells. Eur J Pharmacol 2009, 616, (1-3),
58-63.
113.
Mizuma, T.; Fuseda, N.; Hayashi, M., Kinetic characterization of glycosidase
activity from disaccharide conjugate to monosaccharide conjugate in Caco-2 cells. J
Pharm Pharmacol 2005, 57, (5), 661-664.
114.
Lambert, N.; Kroon, P. A.; Faulds, C. B.; Plumb, G. W.; McLauchlan, W. R.;
Day, A. J.; Williamson, G., Purification of cytosolic beta-glucosidase from pig liver and
its reactivity towards flavonoid glycosides. Bba-Protein Struct M 1999, 1435, (1-2),
110-116.
115.
Henry, C.; Vitrac, X.; Decendit, A.; Ennamany, R.; Krisa, S.; Merillon, J. M.,
Cellular uptake and efflux of trans-piceid and its aglycone trans-resveratrol on the apical
membrane of human intestinal Caco-2 cells. J Agr Food Chem 2005, 53, (3), 798-803.
116.
Higdon, J. V.; Frei, B., Tea catechins and polyphenols: Health effects,
metabolism, and antioxidant functions. Crit Rev Food Sci 2003, 43, (1), 89-143.
31
117.
Rios, L. Y.; Bennett, R. N.; Lazarus, S. A.; Remesy, C.; Scalbert, A.;
Williamson, G., Cocoa procyanidins are stable during gastric transit in humans'. Am J
Clin Nutr 2002, 76, (5), 1106-1110.
118.
Glew, R. H.; Gopalan, V.; Forsyth, G. W.; Vander Jagt, D. J., In: β-
Glucosidases: Biochemistry and Molecular Biology, ACS, Washington, DC. . 1993, 83112.
119.
Lambert, J. D.; Hong, J.; Yang, G. Y.; Liao, J.; Yang, C. S., Inhibition of
carcinogenesis by polyphenols: evidence from laboratory investigations. Am J Clin Nutr
2005, 81, (1), 284s-291s.
120.
Sun, Y.; Peng, Z. L., Programmed cell death and cancer. Postgrad Med J 2009,
85, (1001), 134-140.
121.
Johnstone, R. W.; Ruefli, A. A.; Lowe, S. W., Apoptosis: A link between cancer
genetics and chemotherapy. Cell 2002, 108, (2), 153-164.
122.
Lowe, S. W.; Lin, A. W., Apoptosis in cancer. Carcinogenesis 2000, 21, (3),
485-495.
123.
Thornberry, N. A.; Lazebnik, Y., Caspases: Enemies within. Science 1998, 281,
(5381), 1312-1316.
124.
Kohler, C.; Orrenius, S.; Zhivotovsky, B., Evaluation of caspase activity in
apoptotic cells. J Immunol Methods 2002, 265, (1-2), 97-110.
125.
Vermeulen, K.; Van Bockstaele, D. R.; Berneman, Z. N., Apoptosis:
mechanisms and relevance in cancer. Ann Hematol 2005, 84, (10), 627-639.
126.
Harvey, K. J.; Lukovic, D.; Ucker, D. S., Caspase-dependent Cdk activity is a
requisite effector of apoptotic death events. J Cell Biol 2000, 148, (1), 59-72.
127.
Hampton, M. B.; Fadeel, B.; Orrenius, S., Redox regulation of the caspases
during apoptosis. Towards Prolongation of the Healthy Life Span 1998, 854, 328-335.
128.
Kolenko, V. M.; Uzzo, R. G.; Bukowski, R.; Finke, J. H., Caspase-dependent
and -independent death pathways in cancer therapy. Apoptosis 2000, 5, (1), 17-20.
129.
Boatright, K. M.; Salvesen, G. S., Caspase activation. Proteases and the
Regulation of Biological Processes 2003, 70, 233-242.
130.
Pitti, R. M.; Marsters, S. A.; Ruppert, S.; Donahue, C. J.; Moore, A.; Ashkenazi,
A., Induction of apoptosis by Apo-2 ligand,a new member of the tumor necrosis factor
cytokine family. J Biol Chem 1996, (271), 12687–12690.
32
131.
Ashkenazi, A.; Dixit, V. M., Death receptors: signaling and modulation. .
Science 1998, 281, 1305–1308.
132.
Kim, R.; Emi, M.; Tanabe, K., Caspase-dependent and -independent cell death
pathways after DNA damage (review). Oncol Rep 2005, 14, (3), 595-599.
133.
Zou, H.; Henzel, W. J.; Liu, X. S.; Lutschg, A.; Wang, X. D., Apaf-1, a human
protein homologous to C-elegans CED-4, participates in cytochrome c-dependent
activation of caspase-3. Cell 1997, 90, (3), 405-413.
134.
Adams, J. M.; Cory, S., The Bcl-2 protein family: Arbiters of cell survival.
Science 1998, 281, (5381), 1322-1326.
135.
Mignotte, B.; Vayssiere, J. L., Mitochondria and apoptosis. Eur J Biochem 1998,
252, (1), 1-15.
136.
Cory, S.; Huang, D. C. S.; Adams, J. M., The Bcl-2 family: roles in cell survival
and oncogenesis. Oncogene 2003, 22, (53), 8590-8607.
137.
Mohamad, N.; Gutierrez, A.; Nunez, M.; Cocca, C.; Martin, G.; Cricco, G.;
Medina, V.; Rivera, E.; Bergoc, R., Mitochondrial apoptotic pathways. Biocell 2005,
29, (2), 149-161.
138.
Budd, R. C., Activation-induced cell death. Curr Opin Immunol 2001, 13, (3),
356-362.
139.
Antonsson, B., Mitochondria and the Bcl-2 family proteins in apoptosis
signaling pathways. Mol Cell Biochem 2004, 256, (1-2), 141-155.
140.
Jourdain, A.; Martinou, J. C., Mitochondrial outer-membrane permeabilization
and remodelling in apoptosis. Int J Biochem Cell B 2009, 41, (10), 1884-1889.
141.
O'Connor, L.; Strasser, A.; O'Reilly, L. A.; Hausmann, G.; Adams, J. M.; Cory,
S.; Huang, D. C. S., Bim: a novel member of the Bcl-2 family that promotes apoptosis.
Embo J 1998, 17, (2), 384-395.
142.
Czabotar, P. E.; Colman, P. M.; Huang, D. C. S., Bax activation by Bim? Cell
Death Differ 2009, 16, (9), 1187-1191.
143.
Omelyanchuk, L. V.; Trunova, S. A.; Lebedeva, L. I.; Fedorova, S. A., Key
events of the cell cycle: Regulation and organization. Russ J Genet+ 2004, 40, (3), 219234.
144.
Schafer, K. A., The cell cycle: A review. Vet Pathol 1998, 35, (6), 461-478.
33
145.
Vermeulen, K.; Van Bockstaele, D. R.; Berneman, Z. N., The cell cycle: a
review of regulation, deregulation and therapeutic targets in cancer. Cell Proliferat
2003, 36, (3), 131-149.
146.
Deshpande, A.; Sicinski, P.; Hinds, P. W., Cyclins and cdks in development and
cancer: a perspective. Oncogene 2005, 24, (17), 2909-2915.
147.
Golias, C. H.; Charalabopoulos, A.; Charalabopoulos, K., Cell proliferation and
cell cycle control: a mini review. Int J Clin Pract 2004, 58, (12), 1134-1141.
148.
Morgan, D. O., Principles of Cdk Regulation. Nature 1995, 374, (6518), 131-
134.
149.
Draetta, G., Cell-Cycle Control in Eukaryotes - Molecular Mechanisms of Cdc2
Activation. Trends Biochem Sci 1990, 15, (10), 378-383.
150.
Morgan, D. O., Cyclin-dependent kinases: Engines, clocks, and microprocessors.
Annu Rev Cell Dev Bi 1997, 13, 261-291.
151.
Musgrove, E. A., Cyclins: Roles in mitogenic signaling and oncogenic
transformation. Growth Factors 2006, 24, (1), 13-19.
152.
Pines, J., Cyclins and Cyclin-Dependent Kinases - Theme and Variations.
Advances in Cancer Research, Vol 66 1995, 66, 181-212.
153.
Loyer, P.; Trembley, J. H.; Katona, R.; Kidd, V. J.; Lahti, J. M., Role of
CDK/cyclin complexes in transcription and RNA splicing. Cell Signal 2005, 17, (9),
1033-1051.
154.
Elledge, S. J.; Spottswood, M. R., A New Human P34 Protein-Kinase, Cdk2,
Identified by Complementation of a Cdc28 Mutation in Saccharomyces-Cerevisiae, Is a
Homolog of Xenopus-Eg1. Embo J 1991, 10, (9), 2653-2659.
155.
Doree, M.; Hunt, T., From Cdc2 to Cdk1: when did the cell cycle kinase join its
cyclin partner? J Cell Sci 2002, 115, (12), 2461-2464.
156.
Paterson, L. A.; Meijer, L.; Bentley, M. G., Regulation of M-phase promoting
factor (MPF) in Nereis virens oocytes during meiotic maturation. Invertebr Reprod Dev
1999, 36, (1-3), 175-181.
157.
Liu, F.; Rothblum-Oviatt, C.; Ryan, C. E.; Piwnica-Worms, H., Overproduction
of human Myt1 kinase induces a G(2) cell cycle delay by interfering with the
intracellular trafficking of Cdc2-cyclin B1 complexes. Molecular and Cellular Biology
1999, 19, (7), 5113-5123.
34
158.
Carnero, A.; Hannon, G. J., The INK4 family of CDK inhibitors. Cyclin
Dependent Kinase (Cdk) Inhibitors 1998, 227, 43-55.
159.
Harper, J. W.; Elledge, S. J., Cdk inhibitors in development and cancer. Curr
Opin Genet Dev 1996, 6, (1), 56-64.
160.
Hengst, L.; Reed, S. I., Inhibitors the Cip/Kip family. Cyclin Dependent Kinase
(Cdk) Inhibitors 1998, 227, 25-41.
161.
Brugarolas, J.; Moberg, K.; Boyd, S. D.; Taya, Y.; Jacks, T.; Lees, J. A.,
Inhibition of cyclin-dependent kinase 2 by p21 is necessary for retinoblastoma proteinmediated G(1) arrest after gamma-irradiation. P Natl Acad Sci USA 1999, 96, (3), 10021007.
162.
Gartel, A. L.; Tyner, A. L., Transcriptional regulation of the p21((WAF1/ClP1))
gene. Exp Cell Res 1999, 246, (2), 280-289.
163.
Niculescu, A. B.; Chen, X.; Smeets, M.; Hengst, L.; Prives, C.; Reed, S. I.,
Effects of p21(Cip1/Waf1) at both the G1/S and the G2/M cell cycle transitions: pRb is
a critical determinant in blocking DNA replication and in preventing endoreduplication.
Mol Cell Biol 1998, 18, 629-643.
164.
Radhakrishnan, S. K.; Feliciano, C. S.; Najmabadi, F.; Haegebarth, A.; Kandel,
E. S.; Tyner, A. L.; Gartel, A. L., Constitutive expression of E2F-1 leads to p21dependent cell cycle arrest in S phase of the cell cycle. Oncogene 2004, 23, (23), 41734176.
165.
Gartel, A. L.; Radhakrishnan, S. K., Lost in transcription: p21 repression,
mechanisms, and consequences. Cancer Res 2005, 65, (10), 3980-3985.
166.
O'Reilly, M. A., Redox activation of p21(Cip1/WAF1/Sdi1): A multifunctional
regulator of cell survival and death. Antioxid Redox Sign 2005, 7, (1-2), 108-118.
167.
Xiong, Y.; Hannon, G. J.; Zhang, H.; Casso, D.; Kobayashi, R.; Beach, D., P21
Is a Universal Inhibitor of Cyclin Kinases. Nature 1993, 366, (6456), 701-704.
168.
Waga, S.; Hannon, G. J.; Beach, D.; Stillman, B., The P21 Inhibitor of Cyclin-
Dependent Kinases Controls DNA-Replication by Interaction with Pcna. Nature 1994,
369, (6481), 574-578.
169.
Chavez-Santoscoy, R. A.; Gutierrez-Uribe, J. A.; Serna-Saldivar, S. O., Phenolic
Composition, Antioxidant Capacity and In Vitro Cancer Cell Cytotoxicity of Nine
Prickly Pear (Opuntia spp.) Juices. Plant Food Hum Nutr 2009, 64, (2), 146-152.
35
170.
Zhao, C.; Giusti, M. M.; Malik, M.; Moyer, M. P.; Magnuson, B. A., Effects of
commercial anthocyanin-rich extracts on colonic cancer and nontumorigenic colonic
cell growth. J Agr Food Chem 2004, 52, (20), 6122-6128.
171.
Jourdain, C.; Tenca, G.; Deguercy, A.; Troplin, P.; Poelman, D., In-vitro effects
of polyphenols from cocoa and beta-sitosterol on the growth of human prostate cancer
and normal cells. Eur J Cancer Prev 2006, 15, (4), 353-361.
172.
Yoshida, T.; Hatano, T.; Ito, H., Chemistry and function of vegetable
polyphenols with high molecular weights. Biofactors 2000, 13, (1-4), 121-125.
173.
Al-Ayyoubi, S.; Gali-Muhtasib, H., Differential apoptosis by gallotannin in
human colon cancer cells with distinct p53 status. Mol Carcinogen 2007, 46, (3), 176186.
174.
Lu, J. B.; Ho, C. T.; Ghai, G.; Chen, K. Y., Resveratrol analog, 3,4,5,4 '-
tetrahydroxystilbene, differentially induces pro-apoptotic p53/Bax gene expression and
inhibits the growth of transformed cells but not their normal counterparts.
Carcinogenesis 2001, 22, (2), 321-328.
175.
Hakimuddin, F.; Paliyath, G.; Meckling, K., Selective cytotoxicity of a red grape
wine flavonoid fraction against MCF-7 cells. Breast Cancer Res Tr 2004, 85, (1), 65-79.
176.
Malik, M.; Zhao, C. W.; Schoene, N.; Guisti, M. M.; Moyer, M. P.; Magnuson,
B. A., Anthocyanin-rich extract from Aronia meloncarpa E. induces a cell cycle block
in colon cancer but not normal colonic cells. Nutr Cancer 2003, 46, (2), 186-196.
177.
Tsuchiya, H.; Nagayama, M.; Tanaka, T.; Furusawa, M.; Kashimata, M.;
Takeuchi, H., Membrane-rigidifying effects of anti-cancer dietary factors. Biofactors
2002, 16, (3-4), 45-56.
36
ANTICARCINOGENIC EFFECTS OF POLYPHENOLS FROM DIFFERENT
MANGO (MANGIFERA INDICA L.) VARIETIES
ABSTRACT
Polyphenols found in different cultivars of mango pulp, including gallotannins,
flavonol glycosides, gallic acid, benzophenone derivatives and mangiferin have shown
anticancer activity. The objective of this study was to compare the anti-cancer
properties of phenolic extracts from several mango varieties (Francis, Kent, Ataulfo,
Tommy Atkins and Haden). The antiproliferative effects of mango polyphenols were
studied in vitro using different cancer cell lines including Molt-4 leukemia, A-549 lung,
MDA-MB-231 breast, LnCap prostate, SW-480 colon cancer cells and the non-cancer
colonic myofibroblasts cell line CCD-18Co. Additionally, gene expression was
investigated by RT-PCR and cell cycle kinetics was studied by flow cytometry. Cell
lines were incubated with Ataulfo and Haden phenolic extracts, which were selected
based on their superior antioxidant capacity compared to the other varieties. Ataulfo and
Haden polyphenols inhibited the growth of all human cancer cell lines. SW-480,
MOLT-4 and MDA-MB-231 were statiscally equally most sensitive to Ataulfo, whereas
SW-480 and MOLT-4 were the most sensitive cell lines to Haden, as determined by cell
counting. The efficacy of phenolic extracts from all mango varieties in inhibiting cell
growth was tested on SW-480 colon carcinoma cells. Ataulfo and Haden demonstrated
superior efficacy, followed by Kent, Francis and Tommy Atkins. At 5 mg GAE/L,
Ataulfo inhibited the growth of colon SW-480 cancer cells by ~79% while the growth
of non-cancer colonic myofibroblasts CCD-18Co cells was not inhibited. The growth
inhibition exerted by Ataulfo and Haden polyphenols on SW-480 cells was associated
with an increased mRNA expression of pro-apoptotic biomarkers (caspase 8, Bax and
Bim) and cell cycle regulators (PKMYT1), cell cycle arrest and a decrease in the
generation of reactive oxygen species. Overall, polyphenols from several mango
varieties exerted anti-cancer effects.
Keywords: Mango, Mangifera indica, cancer, cell cycle regulation, polyphenols, gene
expression regulation, cell proliferation.
37
INTRODUCTION
Consumer interest in mango fruits (Mangifera indica) and derived products has
been increasing in recent years based on fruit olfactory properties, and on potential in
preventing chronic diseases (1). Moreover, mango fruits have found their way into the
unofficial classification as so-called superfruit. Cancer is one of the leading chronic
diseases and causes of death worldwide (2). Major types of cancer include lung,
colorectal, breast, and prostate cancer (2). Colon cancer is the third most common
cancer in USA for both men and women, with 108,070 estimated cases of colon and
40,740 cases of rectal cancer diagnosed in 2008 (3). Overall, mortality associated with
colon cancer might be decreased by controlling risk factors. Among these risk factors,
diet is one of the modifiable factors which may be modified through an increased intake
of fruits and vegetables (3).
Plant polyphenols are increasingly being considered as sources of natural colon
cancer chemopreventive compounds based on safety and efficacy assessments (4). The
edible portion of mango contains polyphenols and carotenoids that may protect against
cancer. Polyphenols identified in the edible part of mango have been previously
characterized, and include flavonoids such as quercetin and kaempferol glycosides,
phenolics acids, predominantely gallic acid, hydrolysable tannins, within a wide range
of molecular weight and galloylation, and the xanthone mangiferin (5-6). Bioavailability
of mango polyphenols seems to be limited by high molecular weight and galloylation
degree. However, limited bioavailability of some polyphenols such as gallotannins
might not represent a limitation in the exertion of possible health benefits since they
likely reach the large intestine (7-8), where they may act protecting colon
carcinogenesis.
Few studies have shown the anticancer activity of mango extracts, and they
suggest cell line specificity; e.g. mango extracts have been shown to protect against
prostate cancer in in vitro and in vivo models (9) and to inhibit the cell cycle in the
G(0)/G(1) phase of HL-60 cells (10), although their lack of effectiveness against MCF-7
breast cancer cells has also been reported (11). In addition, some of the phytochemicals
found in mango, i.e. the triterpene lupeol and the xanthone mangiferin have shown to
exibit anti-cancer activity. Mangiferin showed chemopreventive action against lung
38
carcinogenesis induced by benzo(a)pyrene in Swiss albino mice (12). The
chemopreventive action of mangiferin might be related to induction of permeability in
mitochondria (13). Lupeol and related terpenoids exerted varied cytotoxicity against
several cancer cell types (B16 2F2 mouse melanoma, leukemia HL60, U937, K562
melanoma G36; human lung carcinoma (A-549), human colon adenocarcinoma (DLD1) (14).
The overall anti-cancer potential of mango polyphenols might be in part
associated to their antioxidant properties, which contribute to enhance cell defense
capacity and modulate molecular pathways in target cells (15). These compounds may
inhibit promotion and progression stages of cancer by interfering in cell cycle
regulation, signal transduction pathways, transcription and activating apoptosis
(activation of pro-apoptotic genes and pro-apoptotic proteins) in neoplastic cells (16).
The objective of this study was to compare the cancer chemopreventive potential
of polyphenols extracted from different mango varieties in several types of cancer,
while potential molecular mechanisms involved in anti-cancer activities were also
assessed for the most efficacious mango varieties.
MATERIAL AND METHODS
Chemicals
All solvents and chemicals used were of analytical grade. Standards obtained
from Sigma (St. Louis, MO, USA) used for identification purposes by HPLC were as
follows: gallic acid, p-OH-benzoic acid, p-coumaric acid, m-coumaric acid, caffeic acid,
ferulic acid, vannilic acid, mangiferin, quercetin, (+) catechin, ellagic acid and
protocatechuic acid.
Plant material
Commercial varieties of mango (Mangifera indica L.) were kindly donated by
member growers of the National Mango Board. The varieties Francis (Haiti), Kent
39
(Mexico), Ataulfo (Mexico), Tommy Atkins (Mexico), and Haden (Mexico) were
obtained at a firm mid-ripe stage. Fruit were allowed to ripen at room temperature
(23ºC) until subjectively determined to be a full ripe stage as determined by a soft
texture and aroma development. Fruit were manually peeled to remove skin and seeds.
Pulp was homogenized and storage at -80 ºC until needed.
Extraction of polyphenols
Frozen pulp (500 g) was thawed and mixed with 1.5 L of a 1:1:1 (v/v/v) ethanol/
methanol/ acetone solution and filtered with cheesecloth followed by filtration using a
whatman #1 filter paper . The solvents were completely evaporated at 40°C using a
rotavapor (Büchi, Switzerland). The aqueous extract was mixed with methanol (1:1) and
centrifuged at 2000RPM for 10 min at 7°C to remove insoluble precipitates. The
methanol was evaporated in rotavapor at 40 ºC and polyphenols were concentrated on a
C18 cartridge (Waters Corporation, Milford, MA) previously conditioned with acidified
methanol (0.01% HCl) and nanopure water. The aqueous extract was loaded on the C18
cartridge. Polyphenols bound to the matrix were eluted with 50 mL 100% methanol
(F1). The water from the wash (unbound fraction) was partitioned into ethyl acetate
(EtAc) (1:4,v/v) using a separatory funnel in order to recover C18 non-adsorbed
polyphenols, such as phenolic acids. Thus, some hydrophilic non-phenolic compounds
such as organic acids, reducing sugars and soluble proteins were excluded (17). The
ethyl acetate phase (F2) was combined with the methanol eluate (F1). The solvents were
evaporated under vacuum at 40 ºC and the volume made up to 50mL with nanopure
water (Figure 4).
The phenolic aqueous solution was frozen at -80ºC and freeze-dried (Labconco
Corporation, Kansas City, MO) at -50 ˚C and at 0.01mBar of pressure. These extracts
were further used for the cell culture assays by applying different doses based on total
phenolic content measured spectrophotometrically by the Folin-Ciocalteau assay against
an external standard of gallic acid (mg GAE/L) (18). The optical density (OD) values
were recorded on a FLUOstar Omega plate reader (BMG Labtech Inc, Durhan, NC) at
726 nm. Dried extracts were reconstituted in culture medium and sterile filtered prior to
use in cell culture experiments.
40
Crude extract
(ethanol : methanol : acetone)
1:1:1 (v/v/v)
Pulp
(500g)
Solvents
Aqueous extract
Methanol
Pectin
(Methanol)
Aqueous extract
C18 cartridge
Unbound fraction
Elution with methanol
EtAc/Water Partition
Aqueous fraction
(Sugars,
organic acids)
Bound fraction
(F1)
Solvents
Polyphenols
(Phenolic acids)
(F2)
Phenolic extract
(50mL)
Figure 4. Procedure used for extraction of mango polyphenols
Antioxidant capacity (ORAC assay)
The antioxidant capacity of polyphenols from the different mango varieties was
measured using the oxygen radical absorbance capacity (ORAC) assay as described by
Ou et al, 2001 (19), with the use of fluorescein as fluorescent probe. Peroxyl radicals
were generated by 2,2‘-azobis(2-amidinopropane) dihydrochloride, and fluorescence
loss was monitored on a FLUOstar Omega plate reader (BMG Labtech Inc, Durhan,
NC) at 485 nm excitation and 520 nm emission. Each extract was diluted to standardize
the phenolic content up to 10 mg GAE/L in pH 7.2 phosphate buffer prior to pipetting
41
into a 96-well microplate. A 4-fold dilution factor was used in the ORAC assay that
corresponded to an in-well standard concentration ranging from 3.1 to 50 μM Trolox.
Results were quantified in μmol Trolox equivalents/100g pulp.
HPLC-DAD and HPLC-ESI/MSn analysis
Individual polyphenols were characterized using a Waters Alliance 2690
(Milford, MA, USA) HPLC system using a 4.6 x 250 mm Dionex Acclaim 120 C18
column (5µm) and a 4.6 mm x 20 mm guard column. A gradient elution program run
Phase A (water and acetic acid; 98:2) and Phase B (water, acetonitrile, and acetic acid;
68:30:2) at a flow rate of 0.8 mL/min with detection from 210 to 400 nm. The gradient
initially run 100% Phase A for 3 min and changed from 100-70% in 17 min, 70-50% in
10 min, 50-30% in 20 min, 30-0% in 10 min, and then isocratic at 0% Phase A for 5 min
prior to column re-equilibration (20).
Additional identification by mass spectrometric analyses was performed on a
Thermo Finnigan LCQ Deca XP Max MSn ion trap mass spectrometer equipped with
an ESI ion source (Thermo Fisher, San Jose, CA). Polyphenolic extracts were analyzed
using a Waters Alliance 2690 (Milford, MA, USA) HPLC system equipped with a
Dionex Acclaim ® 120 C18 5 µm column, 4.6 mm x 250 mm column and a 4.6 mm x
20 mm guard column. Phenolic compounds were separated using 0.5% formic acid in
water (solvent A) and 0.5% formic acid in acetonitrile (solvent B) as mobile phases, at a
flow rate of 0.4 mL/min. Gradient program ran phase A for 5 min, from 100-70% in 25
min; 70-50% in 15 min; 50-0% in 25 min; and returning to original composition in 8
min.
Ionization was conducted in the negative ion mode under the following
conditions: sheath gas (N2), 40 units/min; auxiliary gas (N2), 5 units/min; spray
voltage, 5 kV; capillary temperature, 350 °C; tube lens offset, 10 V.
Identification of phenolic compounds was performed by comparison of their
retention times and UV-visible spectral characteristics with those from standards
obtained from Sigma-Aldrich (St Louis, MO) as well as their mass spectrometric
properties.
42
Cell culture
Cell lines were obtained from the American Type Culture Collection (ATCC,
Manassas, VA). The estrogen independent MDA-MB-231 breast cancer cells were
cultured using Dulbecco’s modified Eagle’s medium (DMEM) high glucose, with 2
mmol/l L-glutamine, without sodium pyruvate and with phenol red. The colorectal SW480 adenocarcinoma cells, were cultured using Leibovitz`s L-15 medium, with Lglutamine, without sodium pyruvate and phenol red. The human lung A-549 carcinoma
cells were cultured in Dulbecco’s modified Eagle’s medium/F12 (DME/F12). The
human leukemia Molt-4 and the androgen receptor positive LnCap prostate cancer cells
were cultured in RPMI-1640 medium. All culture mediums were supplemented with
10% (v/v) fetal bovine serum (FBS) and 1% Penicillin-Streptomycin antibiotic mix for
propagation. A 2.5% charcoal stripped FBS was used in culture media used for cell
proliferation experiments. Culture media were supplied by Invitrogen (Gibco™,
Invitrogen Corp., Grand Island, NY). Cells were maintained at 37 ºC with a humidified
5% CO2 atmosphere. For cell proliferation assay, cells were seeded in 24 well-plates
(15,000 cells/well), for ROS assay in 96-well plates (10,000 cells/well), for cell cycle
kinetic assay in 12-well plates (70,000 cells/well) and for gene expression studies in 6well plates (500,000 cells/well). Cells were grown for 24 h to allow cell attachment
before exposure to varying concentrations of mango polyphenols (mg GAE/L)
reconstituted in culture medium, and sterile-filtered before use.
Cell proliferation
Cell proliferation was assessed by using an electronic counter (Z2™ Series,
Beckman Coulter, Inc, Fullerton, CA). The difference in cell-number between final
incubation time (72 h) and incubation start (0 h) represents net growth. The cell growth
inhibition was expressed as IC50, the concentration of total polyphenols which inhibits
cell growth by 50%.
43
Cell cycle kinetics
Cells were treated with mango polyphenols (5 and 10 mg GAE/L) for 24 h and
harvested according to Mertens-Talcott et al. (2007) (21). Briefly, cell pellets were
obtained by trypsination and centrifugation. Pellets were fixed with 90% ethanol at -20
°C for 4 h. Cells were resuspended in staining solution [50 µg/mL propidium iodide, 30
units/mL RNase, 4 mmol/L sodium citrate, and Triton X-100 (pH 7.8)] and incubated at
37°C for 15 min. DPBS (Dulbecco's phosphate buffered saline solution) was added to a
final concentration of 0.15 mol/L. Stained cells were analyzed on a FACS Calibur Flow
Cytometer (Becton Dickinson Immunocytometry Systems) using Cell Quest (Becton
Dickinson Immunocytometry Systems) acquisition software as previously described
(21).
Quantitative RT-PCR
Human SW-480 colon adenocarcinoma cells were pretreated with Ataulfo and
Haden polyphenols (2.5-10 mg GAE/L) for 24 h. Total RNA was extracted using the
RNeasy Mini kit (Qiagen, Valencia, CA), and 470 ng of RNA were used to synthesize
cDNA using a Reverse Transcription Kit (Invitrogen Corp., Grand Island, NY). Real
time-PCR reactions were performed using 2µL of cDNA as previously described (22).
Optimal semi-quantitative conditions were set to fall in the linear PCR product range
and TBP (TATA-box binding protein) was used as internal control. The sequences of
the primers (Integrated DNA Technologies, Coralville, IA) used were:
Fas:
F:
5’- TGCCTCCTCTTGAGCAGTCA -3’,
Fas:
R:
5’- TCCTGTAGAGGCTGAGGTGTCA -3’,
Caspase 8:
F:
5’- GGCTCCCCCTGCATCAC-3’,
Caspase 8:
R:
5’- CCTGCTAGATAAGGGCATGAATCT-3’,
Bax:
F:
5’- CCAAGGTGCCGGAACTGA -3’,
Bax:
R:
5’- CCCGGAGGAAGTCCAATGT -3’
Bim:
F:
5’- TGCCAGGCCTTCAACCA -3’,
Bim:
R:
5’- GTTCAGCCTGCCTCATGGA-3’
44
PKMYT1:
F:
5’-CCTCTGCACTTTTAACCTTTTATCCT-3’,
PKMYT1:
R:
5’- GCAGAGAAGACCATGGGAGTTC -3’,
p21:
F:
5’-GAG CTC TGG GTG GTC ATG GA -3’,
p21:
R:
5’- ATC CTG GTG TGG GTG ACG AT -3’,
TBP:
F:
5’-TGCACAGGAGCCAAGAGTGAA-3’,
TBP:
R:
5’-CACATCACAGCTCCCCACCA-3’,
Reactive oxygen species (ROS)
The production of ROS was analyzed according to Meng et al. (2008) (23), with
some modifications. Briefly, cells were pretreated with mango polyphenols (1.5-10mg
GAE/L) over 24 h followed by 200µM H2O2 for 2 h. Upon removal of culture medium
with phenolic extracts, cells were rinsed with DPBS (Dulbecco's phosphate buffered
saline solution, Ca-Mg free), and exposed to 200µM H2O2 for 2h. After washing the
H2O2 with DPBS, cells were exposed to 10µM of 2’, 7’ -dichlorofluorescein diacetate
(DCFH-DA) at 37 °C and incubated for 30min. The fluorescence signal was monitored
at 520 nm emission and 480 nm excitation using a FLUOstar Omega plate reader (BMG
Labtech Inc, Durhan, NC). Fluorescence intensity was used as an indicator of ROS
level.
Statistical analysis
Quantitative data represent mean values of three or more repetitions with the
respective standard deviation (SD) or standard error of the mean (SE). Data were
analyzed by one-way analysis of variance (ANOVA) using post-hoc multiple
comparisons Tukey’s-b (p<0.05 and p<0.01) and Pearson correlations using SPSS
version 12.0 (SPSS Inc., Chicago, IL).
45
RESULTS AND DISCUSSION
Total polyphenols and antioxidant activity of mango varieties
Total phenolic content obtained from mango pulps assessed by Folin-Ciocalteu
assay (Table 1) demonstrated that Ataulfo extracts contained the highest amount of total
polyphenolis normalized to gallic acid equivalent (GAE) (56.7 ± 0.3 mg GAE/100g).
Table 1. Total polyphenols and antioxidant activity (ORAC) of different mango
varieties
Total phenolic content1
Antioxidant activity1
Mango Cultivar
(mg GAE/100g pulp)
(µmolTE/100g pulp)
a
Ataulfo
56.7 ± 0.3
326.6 a ± 4.8
Haden
19.1 b ± 1.8
225.8 b ± 0.6
b,c
Francis
17.8 ± 0.3
219.0 b ± 1.0
Kent
16.4 c ± 0.9
150.0 c ± 0.9
c
156.6 c ± 1.1
Tommy Atkins
15.3 ± 1.6
1
Average of three independent determinations ± SD. Different letters indicates significance (p < 0.05).
Previous studies showed that the total phenolic content from mango pulp may
range from 48.4 to 294 mg GAE/ 100g (24-25), which may be influenced by several
factors including ripening stage, cultivar, geographic production, harvesting, processing
and storage conditions, microbial infection, and so on (5, 25-26). Determination of total
polyphenols might be relevant when analyzing the antioxidant and anticarcinogenic
activities from fruit, since both are in part attributable to the polyphenols content. As
natural antioxidant compounds, polyphenols may interrupt radical chain reactions by
scavenging reactive oxygen species and free radicals (27). Ataulfo phenolic extract was
the highest in antioxidant activity (326.6 ± 4.8 µmol Trolox equivalents/100g mango
pulp), which may be explained by its higher phenolic content as compared to the other
varieties (Table 1). The antioxidant activity of polyphenols from all mango varieties
correlated to their total phenolic content (r = 0.91). In this study, phenolic extracts do
not contain additional antioxidants commonly found in mango including ascorbic acid,
soluble proteins, and reducing sugars. This might explain why previous studies have
been reported higher ORAC values for mango pulp extracts (326-1002 μmol TE/100g),
46
with a slightly higher correlation between ORAC value and total phenolic content (r =
0.98) (24, 28).
HPLC-DAD and HPLC-ESI-MSn analysis of Ataulfo and Haden
polyphenols
The predominant polyphenols in mango pulp were previously identified as gallic
acid and gallotannins, with lower concentrations of numerous other phenolic acids,
flavonol and xanthone glycosides (5-6). Individually, most of the polyphenols present in
mangos contribute to their total antioxidant capacity. The health-related claims of
polyphenols found in mango pulp (29) have been supported by in vitro and in vivo
studies and have been ascribed to their anti-cancer and antioxidant properties (16, 24,
28).
HPLC analysis of Ataulfo and Haden polyphenols confirmed the presence of
gallic acid and mangiferin (Figure 5). Both compounds were found in all cultivars, with
exception of Francis, which did not contain mangiferin (Figure 5). Previous studies also
confirmed our findings (25). Moreover, hydrolyzable tannins composed of gallic acid
units esterified to a glucose moiety (6) were structurally characterized by HPLC-ESIMSn analysis (Table 2, Apendice). Galloyl glucosides identified in Haden presented
chemical structure ranging from one (mono-O-galloyl-glucoside) to nine (nona-Ogalloyl-glucoside) units of gallic acid esterified in a glucose core; whereas the
characterization of Ataulfo phenolic extract revealed the presence of higher molecular
weight gallotannins, varying from mono-O-galloyl-glucoside to undeca-O-galloylglucose. Similarly, gallotannins with 4-7 galloyl residues were previously identified in
mango pulp from the cultivar Tommy Atkins (29). According to Figure 5, polyphenols
with elution pattern after 58 min comprise gallotannins with molecular weight ranging
from 939 (penta-O-galloyl-glucoside) to 1851 Da (undeca-O-galloyl-glucose).
47
3.00
0.035
(A)
1
0.030
3
(A1)
0.025
AU
0.020
0.015
0.010
2.00
0.005
AU
0.000
25.00
30.00
35.00
40.00
Minutes
45.00
50.00
1
1.00
1
1
1
1
2
1
55.00
11 11
1
1
1
1
11 1
11
0.00
10.00
0.10
20.00
30.00
(B)
40.00
Minutes
1
50.00
0.006
0.004
60.00
70.00
5
3
(B11)
AU
0.002
4
0.00
0.000
-0.002
-0.004
AU
10.00
1
1
-0.20
20.00
3
-0.10
1
1
30.00
40.00
Minutes
50.00
1
1111
1
1
60.00
70.00
1
1
1
1 2
1
1
1
3
-0.30
10.00
20.00
30.00
40.00
Minutes
50.00
60.00
70.00
Figure 5. Representative chromatograms at 280 nm of Ataulfo (A) and Haden
polyphenols (B). (1) gallotannins; (2) gallic acid. Representative chromatograms at 366
nm of Ataulfo (A1) and Haden (B1) (3) mangiferin.
48
Table 2. Polyphenols profile from the mango varieties Ataulfo, Haden, Kent, Francis,
and Tommy Atkins determined by HPLC-DAD and HPLC-ESI-MSn analysis.
Class
Phenolic acid
Xanthone
Gallotannins
Polyphenols
Ataulfo
Haden
Kent
Francis
Gallic acid
mangiferin
9
9
9
9
9
9
9
nd
mono-O-galloyl- glucose
9
9
9
9
di-O-galloyl- glucose
nd
9
tetra-O-galloyl- glucose
nd
9
9
9
nd
nd
Tommy
Atkins
9
9
9
nd
nd
penta-O-galloyl- glucose
9
9
9
9
9
hexa-O-galloyl- glucose
9
9
9
9
9
hepta-O-galloyl- glucose
9
9
9
9
9
octa-O-galloyl- glucose
9
9
9
9
9
nona-O-galloyl- glucose
9
9
9
9
9
deca-O-galloyl- glucose
9
nd
nd
nd
nd
undeca-O-galloyl-glucose
9
nd
nd
nd
nd
nd= not determined or not detected.
Cell-growth suppressive activity of Haden and Ataulfo mango
polyphenols on different cancer cell lines
Based on their superior antioxidant capacity, Ataulfo and Haden were selected to
assess the growth inhibition activity of their polyphenols in different cancer cell lines
(Figure 6).
49
120
80
120
LnCap
Molt-4
100
A-549
MDA-MB-231
80
SW-480
60
60
(A)
Net Growth (% of control)
Net Growth (% of control)
100
40
20
0
-20
(B)
LnCap
Molt-4
A-549
MDA-MB-231
SW-480
40
20
0
-20
-40
0
10
20
30
-40
50 0
40
Ataulfo polyphenolics (mg GAE/L)
10
20
30
40
50
Haden polyphenolics (mg GAE/L)
Figure 6. Cell proliferation of prostate LnCap, leukemia Molt-4, lung A-549, breast
MDA-MB-231 and colon SW-480 cancer cells treated with Ataulfo (A) and Haden (B)
polyphenols. Cells were treated with different concentrations of extracts and cell growth
was assessed after 72 h incubation. Values are means ± SD, n=3.
Ataulfo and Haden polyphenols inhibited the cell proliferation of all cancer cell
lines in a concentration-dependent manner. The IC50 values (concentration that inhibits
the growth by 50%) show that Ataulfo polyphenols suppressed the cancer cell growth in
the following order of efficacy: SW-480 = Molt-4 = MDA-MB-231 > A-549 = LnCap
within a range of 0-42 mg GAE/L (Table 3). Likewise, IC50 values indicate that human
colorectal SW-480 carcinoma cells were inhibited by both Haden and Ataulfo
polyphenols to a similar extent (Table 3).
Table 3. IC50 values of polyphenols extracted from Ataulfo and Haden mango varieties
for growth suppression of different human cancer cell lines
Mango
Cultivar
IC50 (mg GAE/L)
SW-480
Ataulfo 1.61 ± 0.2b
Haden
2.31 ± 0.2b
MDA-MBLnCap
A-549
Molt-4
231
1.31 ± 0.1b 18.02 ± 0.0a 13.21 ± 5.5a 5.01 ± 1.0b
> 10 2
8.31 ± 0.0a
ND
7.01 ± 0.0a
1
8.32 ± 1.8a
2.01 ± 0.5b
CCD-18Co
Average of three independent determinations ± SD. 2Average of two independent determinations.
Different letters indicate statistical significance at p< 0.05. ND, not determined.
50
Overall, mango polyphenols have low bioavailability, in part, due to the
presence of higher molecular weight polyphenols such as gallotannins (8, 30-31). Thus,
the chemopreventive protection of mango polyphenols, specifically those with larger
molecular weight, may be relevant to the prevention of colon cancer in vivo. However,
limited bioavailability of high molecular weight polyphenols might not represent a
limitation in the exertion of possible health benefits since they likely reach the large
intestine (7-8), where they may act protecting colon carcinogenesis.
Overall, these results suggest that polyphenols from Ataulfo and Haden varieties
inhibit the proliferation of all tested types of cancer cell lines, and SW-480 colon cancer
cell line had high susceptibility to the treatment.
Growth suppressive activity of mango polyphenols from different
varieties on colon cancer cells
Based on their high sensitivity to mango polyphenols and the likely in vivo
effects of mango polyphenols in modulating human gut microflora, SW-480 cells were
selected for the following analyses. Polyphenols extracted from Ataulfo, Haden, Kent,
Francis and Tommy Atkins induced a concentration dependent growth-suppression in
human SW-480 colon cancer cells (Figure 7).
51
Net Growth of SW-480 colon
cancer cells (% of control)
120
Kent
Tommy Atkins
Haden
Ataulfo
Francine
100
80
60
40
20
0
-20
-40
0
5
10
15
20
Polyphenolics (mg GAE/L)
Figure 7. Cell-growth suppressive effects of polyphenols from Kent, Tommy Atkins,
Haden, Ataulfo and Francis mango varieties on colon SW-480 human cancer cells. Cells
were incubated with extracts and net growth was measured after 72 h incubation. Values
are mean ± SD, n=3.
Among the five cultivars, Ataulfo and Haden polyphenols showed the highest
cell growth-inhibition in SW-480. At 1.3 and 2.5 mg GAE/L, Ataulfo polyphenols
suppressed the growth of SW-480 cells by 36% and 65%, respectively, at 72 h of
incubation. When incubated with 5 mg GAE/L of Ataulfo polyphenols, net cell growth
was decreased by 22% indicating that the number of cells at 72h was lower than at the
baseline (0 h), suggesting net cell killing (Figure 7). Polyphenols from Haden reduced
cell growth in a concentration-dependent manner, with 63% and 89% of inhibition
achieved at 2.5 and 5 mg GAE/L respectively. The order of efficacy for the tested
mango extracts was: Ataulfo = Haden ≥ Kent ≥ Francis > Tommy Atkins based on their
IC50 values (Table 4).
52
Table 4. IC50 values of polyphenols extracted from mango varieties for growth
suppression of human SW-480 colon cancer cells
1
Mango Cultivar
Ataulfo
Haden
Kent
Francis
Tommy Atkins
IC50 (mg GAE/L)
1.61 ± 0.2a
2.31 ± 0.2a
5.02 ± 1.4a,b
8.21 ± 2.6b
27.32 ± 0.4c
Average of three independent determinations ± SD. 2Average of two independent determinations.
Different letters indicate statistical significance at the level p< 0.05.
A previous study reported low cytotoxic efficacy of mango aqueous extracts in
MCF-7 breast cancer cells when compared to other plant-based foods consumed in
Mexico (11). Polyphenols in that study were extracted with water and did not contain
compounds with lower solubility in water such as higher molecular weight gallotannins,
as demonstrated by Percival et al., 2006 (10).
Apparently, the degree of galloylation may play a role in the cytotoxic effects in
cancer cells (32-33), which may indicate that some of the anti-cancer effects may be
increased by the high degree of galloylation of mango-polyphenols. Tannin structureactivity relationship studies indicate that tannins which exhibited potent inhibitory
activity on the tumor cell motility have common characteristics such as galloyl groups
substituting all the hydroxyl groups of β-D-glucose and some of them cross-linked to
form hexahydroxydiphenoyl. Ellagitannins including the most potent 1,2,4-tri-Ogalloyl-3,6-hexahydroxydiphenoyl-β-D-glucose (punicafolin) showed to be more potent
than gallotannins (mono, di and penta-O-galloyl-β-D-glucose) (34). In similar study,
Coriariin A, which has four galloyl groups, showed higher antitumor effects than
agrimoniin, that has two hexahydroxydiphenol groups (35). This results are in
agreement with Lizarra et al. (2008) (33) findings. The more galloylated Witch hazel
fractions showed better effectiveness at inhibiting proliferation of HT-29 and HCT-116
human colon cancer cells, at arresting the cell cycle at the S phase as well as at inducing
apoptosis and necrosis. Interestingly, the apoptosis and cell cycle arrest effects were
proportional to their galloylation level. Moreover, witch hazel fractions with a high
degree of galloylation were also the most effective as scavengers of both hydroxyl and
superoxide radicals and in protecting DNA damage triggered by the hydroxyl radical
system. Moreover, tannin-rich fraction (85.1- 93.2% of the TPC) extracted from four
53
cultivars of muscadine grapes showed a 2-4 greater inhibitory activity against HT-29
and Caco-2 colon cancer cells, when compared to phenolic acid fraction (36), likely as a
result of its higher capacity in preventing oxidative stress (37-38).
In summary, SW-480 colon cancer cells were affected by the treatment with the
selected mango polyphenols and phenolic extracts from Ataulfo and Haden were
assessed as being most efficacious in the induction of cell death when compared to other
mango varieties.
Growth-suppressive activity of Ataulfo polyphenols on cancer cells as
compared to normal cells
The growth inhibition effects of mango polyphenols were investigated in noncancerous colonic myofibroblasts (CCD-18Co) cells compared to SW-480 cells. At 5
mg GAE/L, Ataulfo polyphenols suppressed the growth of SW-480 cells by ~ 79 %,
whereas the growth of CCD-18Co cells was not inhibited at the same concentration.
Even at the highest concentration of Ataulfo polyphenols (10 mg GAE/L), the growth
inhibition of CCD-18Co cells was only 13 ± 6% (Figure 8). Previous studies have
reported that growth-inhibitory effects of polyphenols were greater in colon cancer as
compared to non-tumorigenic colon cells (39-40). In general, bioactive natural
compounds which has growth-suppressive effects in cancer cells but do not affect
normal cells, may have a potential in chemoprevention, which is suggested by these
data.
54
CCD-18Co
SW-480
Net Growth (% of control)
120
100
80
60
40
20
0
0
2
4
6
8
10
12
Ataulfo polyphenols (mg GAE/L)
Figure 8. Cell-growth suppressive effects of Ataulfo polyphenols on human SW-480
colon cancer cells and colonic myofibroblasts CCD-18Co cells. Cells were incubated
with phenolic extract and net growth was measured after 48 h. Values are mean ± SD,
n=3. Asterisk indicates a significant difference compared to untreated control (*) p ≤
0.01.
Cell Cycle Regulation
Ataulfo and Haden polyphenols induced G2/M cycle arrest (Figure 9). Haden
polyphenols at 10 mg GAE/L were more effective than Ataulfo in increasing the
number of cells in G2/M phase by 1.9-fold. Haden polyphenols also induced cell cycle
arrest in the S phase in a dose-dependent manner, whereas Ataulfo polyphenols did not
show significant effect at this phase. Previous studies demonstrated that polyphenols
from mango influenced cell cycle transition in Caco-2 colon cancer cells with an
increased percentage of cells in the S and G2/M phase (41). Similarly, grape
polyphenols with a high percentage of galloylation showed to be effective in arresting
the cell cycle in the G2/M phase and inducing apoptosis in HT29 human colon cancer
cells (32). Gallotannins induced S-phase arrest in HCT-116 human colon cancer cells,
which was associated with an increase in p53 protein levels and p21 mRNA and protein
levels (42). Overall, the demonstrated cell cycle modulating properties of gallic acid and
55
galloylated derivatives as well as other polyphenols present in mango pulp have likely
contributed to the anti-cancer properties in SW-480 cancer cells.
Percentage of cells in Cell Cycle Phase (%)
60
(A)
a
G0
S
G2/M
b, c
50
c
40
30
a
a
20
b
10
0
-5
0
5
10
15
Ataulfo
polyphenols
(mg
GAE/L)
Ataulfo
Ataulfo
polyphenolics
polyphenolics
(mg
GAE/L)
Percentage of cells in Cell Cycle Phase (%)
70
G0
S
G2/M
(B)
a
60
b
50
a
c
40
b
c
30
a,b
20
b,c
c
10
0
-5
0
5
10
15
Hadenpolyphenolics
polyphenols (mg
Haden
(mgGAE/L)
GAE/L)
Figure 9. Cell cycle analysis of SW-480 colon cancer cells treated with Ataulfo and
Haden polyphenols for 24 h. Values are means ± SE (n=3), different letters indicate
significance at p < 0.05.
56
Gene Transcriptional Regulation
The expression of genes involved in the regulation of apoptosis, referred to as
“programmed cell death” were investigated. Two pathways involved in the activation of
caspases have been studied: the extrinsic death receptor pathway induced by ligation of
the Fas-receptors, or Fas-mediated apoptosis, and the intrinsic or mitochondrial pathway
(43). The former leads to activation of caspase-8, an initiator of the caspase cascade
resulting in proteolysis of cellular proteins and death by apoptosis (43), whereas the
latter is mainly regulated by the Bcl-2 family members, which include pro-apoptotic and
pro-survival proteins (44). Some pro-apoptotic proteins, i.e. Bax form channels and
contribute to regulate preexisting channels that permeabilize the outer mitochondrial
membrane, which releases caspase-activating proteins into the cytosol (45), whereas
others i.e. Bim neutralize certain pro-survival Bcl-2 sub-family of proteins (46).
The effects of Haden and Ataulfo polyphenols on the expression of genes
involved in apoptosis were investigated within a concentration-range 0-10 mg GAE/L in
SW-480 cells. Results indicate that both the extrinsic death receptor and the intrinsic
mitochondria pro-apoptotic pathways were targeted at the transcriptional level by
Ataulfo and Haden polyphenols, after 24 h of treatment (Figure 10).
Caspase 8 gene expression was up-regulated by Ataulfo at the highest dose
(10mg GAE/L) by 2-fold (Figure 10.A), suggesting the contribution of the extrinsic
pathway to the induction of apoptosis. However, the expression of Fas did not seem to
be regulated within the time frame and concentration range used and/or might be being
targeted at post-translational level (43). When used at the highest concentrations,
Ataulfo polyphenols up-regulated the transcription of mitochondria-related proapoptotic Bax and Bim by 1.3 and 1.7 fold respectively (Figure 10.A), whereas Haden
polyphenols only increased Bax mRNA (2.6 fold), without any effect on Bim gene
expression (Figure 10.B).
57
(A)
Fas
3
a
a
a
1
2.5
2.0
2.5
5
a
a
a
1
0
Bax
b
a
a
a
1.0
0.5
3
2
2.5
5
10
Ataulfo polyphenols (mg GAE/L)
Bim
b
a
a
a
1
0
0
2.5
5
10
Ataulfo polyphenols (mg GAE/L)
mRNA levels/ratio toTBP
b
2
10
Ataulfo polyphenols (mg GAE/L)
1.5
0.0
2.5
Caspase 8
3
0
0
mRNA levels/ratio toTBP
2
0
3.0
mRNA levels/ratio toTBP
mRNA levels/ratio toTBP
a
b
2.0
a, b
a,b
1.5
a
1.0
0.5
0.0
0
p21
mRNA levels/ratio toTBP
mRNA levels/ratio toTBP
4
3
2.5
5
10
Ataulfo polyphenols (mg GAE/L)
PKMYT1
b
2
a, b
a
a
1
0
0
2.5
5
0
10
2.5
5
10
Ataulfo polyphenolics (mg GAE/L)
Ataulfo polyphenolics (mg GAE/L)
58
Fas
3
a
a
2
a
a
1
a
1
10
Haden polyphenols (mg GAE/L)
b
a
a
a
1
0
2.5
5
mRNA levels/ratio toTBP
1.5
a
a
a
a
1.0
0.5
0.0
2.0
2.5
5
10
Haden polyphenols (mg GAE/L) Bim
a
a
1.5
a
a
1.0
0.5
0.0
5
10
Haden polyphenols (mg GAE/L)
2.0
0
Bax
mRNA levels/ratio toTBP
5
p21
mRNA levels/ratio toTBP
mRNA levels/ratio toTBP
2.5
3
0
2.5
a, b
a, b
0
0
2
b
2
0
4
Caspase 8
3
mRNA levels/ratio toTBP
mRNA levels/ratio toTBP
(B)
0
2.5
5
10
PKMYT1
Haden polyphenols (mg GAE/L)
4
b
3
2
a
a
a
2.5
5
1
0
0
2.5
5
10
0
Haden polyphenolics (mg GAE/L)
10
Haden polyphenolics (mg GAE/L)
Figure 10. mRNA expression of SW-480 colon cancer cells treated with Ataulfo (A)
and Haden (B) polyphenols after 24 h and analyzed by real time PCR as ratio to TATAbinding protein (TBP) mRNA. Values are means ± SE (n=3). Different letters indicate
significance at p < 0.05.
Natural plant extracts and phytochemicals appear to target the intrinsic
mitochondrial pathway (13, 47) and a crosstalk between the intrinsic and extrinsic
pathways occurs (45). This is the first study reporting gene expression regulation of
mango polyphenols on colon SW-480 cancer cells. However the main polyphenols
59
identified in mango, i.e. gallotannins have shown to regulate the pro-apototic and cell
cycle related proteins on colon (42) and breast cancer cells (48). The structurally related
ellagitannins also regulated the expression of cell cycle and genes of the Bcl-2 family
members in MDA-MB-231 breast cancer cells (49).
In this study, the effects of mango polyphenols on the expression of genes
encoding the cell cycle regulator proteins p21 (Cip1/WAF1) (p21) and the protein
kinase-membrane associated tyrosine/threonine 1 (PKMYT1), involved were analyzed.
Results showed that Ataulfo and Haden polyphenols do not affect the expression of p21
(Cip1/WAF1) mRNA (Figure 10), which was consistent with no regulation of G0/G1
phase for Ataulfo treated-cells, but not consistent with the observed inhibition in G0/G1
phase for cells treated with Haden polyphenols (Figure 9.A). Both Ataulfo and Haden
polyphenols upregulated PKMYT1 gene expression by 1.4 and 3 fold, respectively. The
protein encoded by PKMYT1 gene is a member of the serine/threonine protein kinase
family. This kinase preferentially phosphorylates and inactivates cell division cycle 2
protein (CDC2), and thus supress G2/M transition, which result in G2/M arrest (50).
Individual phytochemicals found in mango have been reported to modulate
genes involved in cell cycle regulation, i.e. quercetin down-regulated cyclin D and
survivin at both the transcription and protein expression levels in SW480 colon cancer
cells (51); penta-O-galloyl-β-D-glucose induced cell cycle arrests in the S and G(1)
phases on prostate cancer cells (52), and gallic acid decreased the expression of Cdks in
vivo (53). As observed with many individual polyphenols, the influence on gene
expression by mango polyphenols as a whole in SW-480 colon cancer cells indicates
that growth inhibition is at least in part mediated by the regulation of pro-apoptotic
genes and cell cycle control genes.
Protective effects against reactive oxygen species (ROS)
Oxidative damage by reactive oxygen species seems to be a crucial event for the
initiation of cancer (15), and a chemopreventive agent might interfere with this step and
prevent cancer by reducing the ROS production. Therefore the potential of mango
polyphenols in protecting the colon cells against reactive oxygen species (ROS) was
investigated. Pre-treatment of SW-480 cells with 1.5, 2.5 and 5 mg GAE/L Ataulfo
60
polyphenols for 24h significantly reduced the 200 µM H2O2 induced-ROS production
by ~ 14%, 28% and 70%. Haden polyphenols were less effective in protecting against
H2O2-induced oxidative damage at 2.5 and 5 mg GAE/L polyphenols, with 14% and
17% reduction of ROS production compared to the control (Figure 11.A). However, the
concentration-dependent protection of SW-480 against ROS production was reversed at
the highest dose (10mg GAE/L) by both Ataulfo and Haden polyphenols.
Previous findings suggest that cancer cells use ROS signals to drive proliferation
and other events required for tumor progression (54). Therefore the ROS generation
inhibition may contribute to slow down the tumor progression. The observed increase of
ROS in cancer cells caused by mango polyphenols at higher concentrations may have
been caused by an increased basal oxidative stress which may have driven them beyond
a tumor sustaining threshold, making them more sensitive to oxidative stress that finally
ends in cell death (54).
In contrast, when the non-cancer colonic myofibroblasts CCD-18Co cells where
pre-treated with Ataulfo and Haden polyphenols at the same concentration-range (1.510 mg GAE/L), a protection against ROS production was achieved in a dose-dependent
manner, including the highest concentration of 10mg/L. Within 1.5-10 mg GAE/L,
Ataulfo and Haden reduced the ROS generation by 14-25% and 6-16%, respectively
(Figure 11B). These results demonstrate that at concentrations which induce oxidative
damage in cancer cells (10mg GAE/L), mango polyphenols still protect the non-cancer
cells against ROS. Overall, Ataulfo polyphenols were more potent compared to Haden
in the protection of non-cancer colon CCD-18Co cells against ROS. These results
corroborate previous findings which demonstrated how natural compounds may target
the production of ROS in their cytotoxic effects in cancer cells but not in normal cells
(55).
61
120
Ataulfo
Haden
c
(A)
Percentage of control (%)
a a
a
100
b
b
80
b
c
c
60
40
d
20
0
0
120
1.5
5
10
Polyphenolics (mg GAE/L)
(B)
Ataulfo
Haden
a a
Percentage of control (%)
2.5
100
b
b
b,c
b,c
b,c
b,c
c
c
80
60
40
20
0
0
1.5
2.5
5
10
Polyphenolics (mg GAE/L)
Figure 11: Protective effects of Ataulfo and Haden polyphenols against H2O2- induced
ROS production on SW-480 cancer cells (A) and CCD-18Co non-cancer cells (B). Cells
were pretreated with extract for 24h and exposed to 200μM H2O2 for 2h. Values are
means ± SD (n=6), different letters indicate significance at p < 0.05.
Conclusion
Polyphenols identified in the edible part of these mango varieties comprise gallic
acid, mangiferin and a wide range of high molecular weight hydrolysable tannins.
Ataulfo and Haden polyphenols inhibited the growth of human SW-480 colon cancer
62
cells which were showed high sensitivity to the treatment. SW-480 gene regulation
influenced by Ataulfo and Haden polyphenols suggested the induction of apoptosis
through intrinsic and extrinsic mechanisms, in addition to cell cycle arrest. Likewise,
cell cycle arrest in the G2/M phase was related to the upregulation of PKMYT1 gene
expression. Ataulfo and Haden polyphenols exerted protection of non-cancer CCD18Co colon cells by lowering the ROS generation in a dose dependent manner. Based
on their anti-cancer effects and protective effects in normal cells, but not in cancer cells,
mango polyphenols may have a high potential as chemopreventive agents.
63
REFERENCES
1.
Masibo, M.; He, Q., Major mango polyphenols and their potential significance
to human health. Compr Rev Food Sci F 2008, 7, (4), 309-319.
2.
WHO, World Health Statistics 2008, World Health Organization: Geneva
Switzerland, 2008.
3.
American
Cancer
Society
Cancer
Facts
&
Figures
2008
http://www.cancer.org/downloads/STT/2008CAFFfinalsecured.pdf (03/13/09).
4.
Rudolf, E.; Andelova, H.; Cervinka, M., Polyphenolic compounds in
chemoprevention of colon cancer - targets and signaling pathways. Anticancer Agents
Med Chem 2007, 7, (5), 559-75.
5.
Schieber, A.; Ullrich, W.; Carle, R., Characterization of polyphenols in mango
puree concentrate by HPLC with diode array and mass spectrometric detection
Innovative Food Science and Emerging Technologies 2000, 1, (2), 161-166.
6.
Barreto, J. C.; Trevisan, M. T. S.; Hull, W. E.; Erben, G.; de Brito, E. S.;
Pfundstein, B.; Wurtele, G.; Spiegelhalder, B.; Owen, R. W., Characterization and
quantitation of polyphenolic compounds in bark, kernel, leaves, and peel of mango
(Mangifera indica L.). J Agr Food Chem 2008, 56, (14), 5599-5610.
7.
Scalbert, A.; Manach, C.; Morand, C.; Remesy, C., Bioavailability of dietary
polyphenols is a key issue to assess their impact on physiological functions. Free
Radical Bio Med 2004, 36, S27-S27.
8.
Scalbert, A.; Williamson, G., Dietary intake and bioavailability of polyphenols.
Journal of Nutrition 2000, 130, (8), 2073s-2085s.
9.
Prasad, S.; Kalra, N.; Shukla, Y., Induction of apoptosis by lupeol and mango
extract in mouse prostate and LNCaP cells. Nutr Cancer 2008, 60, (1), 120-30.
10.
Percival, S. S.; Talcott, S. T.; Chin, S. T.; Mallak, A. C.; Lounds-Singleton, A.;
Pettit-Moore, J., Neoplastic transformation of BALB/3T3 cells and cell cycle of HL-60
cells are inhibited by mango (Mangifera indica L.) juice and mango juice extracts.
Journal of Nutrition 2006, 136, (5), 1300-1304.
11.
Garcia-Solis, P.; Yahia, E. M.; Morales-Tlalpan, V.; Diaz-Munoz, M., Screening
of antiproliferative effect of aqueous extracts of plant foods consumed in Mexico on the
breast cancer cell line MCF-7. Int J Food Sci Nutr 2009, 1-15.
64
12.
Rajendran, P.; Ekambaram, G.; Sakthisekaran, D., Effect of mangiferin on
benzo(a)pyrene induced lung carcinogenesis in experimental Swiss albino mice. Natural
Product Research 2008, 22, (8), 672-680.
13.
Pardo-Andreu, G. L.; Dorta, D. J.; Delgado, R.; Cavalheiro, R. A.; Santos, A. C.;
Vercesi, A. E.; Curti, C., Vimang (Mangifera indica L. extract) induces permeability
transition in isolated mitochondria, closely reproducing the effect of mangiferin,
Vimang's main component. Chem-Biol Interact 2006, 159, (2), 141-148.
14.
Chaturvedi, P. K.; Bhui, K.; Shukla, Y., Lupeol: Connotations for
chemoprevention. Cancer Letters 2008, 263, (1), 1-13.
15.
Russo, G. L., Ins and outs of dietary phytochemicals in cancer chemoprevention.
Biochemical Pharmacology 2007, 74, (4), 533-544.
16.
Surh, Y. J., Cancer chemoprevention with dietary phytochemicals. Nat Rev
Cancer 2003, 3, (10), 768-780.
17.
Medlicott, A. P.; Thompson, A. K., Analysis of Sugars and Organic-Acids in
Ripening Mango Fruits (Mangifera-Indica L Var Keitt) by High-Performance LiquidChromatography. J Sci Food Agr 1985, 36, (7), 561-566.
18.
Swain, T.; Hillis, W. E., The phenolic constituents of Prunus domestica I. The
quantitative analysis of phenolic constituents. Journal of Science Food Agricultural
1959, 10, 63-68.
19.
Ou, B. X.; Hampsch-Woodill, M.; Prior, R. L., Development and validation of
an improved oxygen radical absorbance capacity assay using fluorescein as the
fluorescent probe. J Agr Food Chem 2001, 49, (10), 4619-4626.
20.
Talcott, S. T.; Howard, L. R.; Brenes, C. H., Contribution of periderm material
and blanching time to the quality of pasteurized peach tree. J Agr Food Chem 2000, 48,
(10), 4590-4596.
21.
Mertens-Talcott, S. U.; De Castro, W. V.; Manthey, J. A.; Derendorf, H.;
Butterweck, V., Polymethoxylated flavones and other phenolic derivates from citrus in
their inhibitory effects on P-glycoprotein-mediated transport of talinolol in Caco-2 cells.
J Agr Food Chem 2007, 55, (7), 2563-2568.
22.
Mertens-Talcott, S. U.; Chintharlapalli, S.; Li, M. R.; Safe, S., The oncogenic
microRNA-27a targets genes that regulate specificity protein transcription factors and
65
the G(2)-M checkpoint in MDA-MB-231 breast cancer cells. Cancer Research 2007,
67, (22), 11001-11011.
23.
Meng, Q. Y.; Velalar, C. N.; Ruan, R. S., Effects of epigallocatechin-3-gallate
on mitochondrial integrity and antioxidative enzyme activity in the aging process of
human fibroblast (vol 44, pg 1032, 2008). Free Radical Bio Med 2008, 45, (12), 17381738.
24.
Kim, Y.; Brecht, J. K.; Talcott, S. T., Antioxidant phytochemical and fruit
quality changes in mango (Mangifera indica L.) following hot water immersion and
controlled atmosphere storage. Food Chem 2007, 105, (4), 1327-1334.
25.
Ribeiro, S. M. R.; Barbosa, L. C. A.; Queiroz, J. H.; Knodler, M.; Schieber, A.,
Phenolic compounds and antioxidant capacity of Brazilian mango (Mangifera indica L.)
varieties. Food Chem 2008, 110, (3), 620-626.
26.
Rocha, R. S. M.; Queiroz, J. H.; Lopes, R. Q. M. E.; Campos, F. M.; Pinheiro, S.
H. M., Antioxidant in mango (Mangifera indica L.) pulp. Plant Foods Hum Nutr. 2007,
62, (1), 13-17.
27.
Shahidi, F.; Janitha, P. K.; Wanasundara, P. D., Phenolic Antioxidants. Crit Rev
Food Sci 1992, 32, (1), 67-103.
28.
Wu, X. L.; Beecher, G. R.; Holden, J. M.; Haytowitz, D. B.; Gebhardt, S. E.;
Prior, R. L., Lipophilic and hydrophilic antioxidant capacities of common foods in the
United States. J Agr Food Chem 2004, 52, (12), 4026-4037.
29.
Berardini, N.; Carle, R.; Schieber, A., Characterization of gallotannins and
benzophenone derivatives from mango (Mangifera indica L. cv. 'Tommy Atkins') peels,
pulp and kernels by high-performance liquid chromatography electrospray ionization
mass spectrometry. Rapid Commun Mass Sp 2004, 18, (19), 2208-2216.
30.
Coates, E. M.; Popa, G.; Gill, C. I. R.; McCann, M.; McDougall, G. J.; Stewart,
D.; Rowland, I., Colon-available raspberry extract exhibit anti-cancer effects on in vitro
models of colon cancer. P Nutr Soc 2007, 66, 67a-67a.
31.
Carbonaro, M.; Grant, G.; Pusztai, A., Evaluation of polyphenol bioavailability
in rat small intestine. Eur J Nutr 2001, 40, (2), 84-90.
32.
Lizarraga, D.; Lozano, C.; Briede, J. J.; van Delft, J. H.; Tourino, S.; Centelles,
J. J.; Torres, J. L.; Cascante, M., The importance of polymerization and galloylation for
66
the antiproliferative properties of procyanidin-rich natural extracts. Febs J 2007, 274,
(18), 4802-11.
33.
Lizarraga, D.; Tourino, S.; Reyes-Zurita, F. J.; de Kok, T. M.; van Delft, J. H.;
Maas, L. M.; Briede, J. J.; Centelles, J. J.; Torres, J. L.; Cascante, M., Witch Hazel
(Hamamelis virginiana) Fractions and the Importance of Gallate Moieties-Electron
Transfer Capacities in Their Antitumoral Properties. J Agr Food Chem 2008, 56, (24),
11675-11682.
34.
Tanimura, S.; Kadomoto, R.; Tanaka, T.; Zhang, Y. J.; Kouno, I.; Kohno, M.,
Suppression of tumor cell invasiveness by hydrolyzable tannins (plant polyphenols) via
the inhibition of matrix metalloproteinase-2/-9 activity. Biochem Bioph Res Co 2005,
330, (4), 1306-1313.
35.
Miyamoto, K.; Kishi, N.; Koshiura, R.; Yoshida, T.; Hatano, T.; Okuda, T.,
Relationship between the Structures and the Antitumor Activities of Tannins. Chem
Pharm Bull 1987, 35(2) (2), 814-822.
36.
Yi, W. G.; Fischer, J.; Akoh, C. C., Study of anticancer activities of muscadine
grape phenolics in vitro. J Agr Food Chem 2005, 53, (22), 8804-8812.
37.
Osawa, T., Protective role of dietary polyphenols in oxidative stress.
Mechanisms of Ageing and Development 1999, 111, (2-3), 133-139.
38.
Urquiaga, I.; Leighton, F., Plant polyphenol antioxidants and oxidative stress.
Biol Res 2000, 33, (2), 55-64.
39.
Chavez-Santoscoy, R. A.; Gutierrez-Uribe, J. A.; Serna-Saldivar, S. O., Phenolic
Composition, Antioxidant Capacity and In Vitro Cancer Cell Cytotoxicity of Nine
Prickly Pear (Opuntia spp.) Juices. Plant Food Hum Nutr 2009, 64, (2), 146-152.
40.
Zhao, C.; Giusti, M. M.; Malik, M.; Moyer, M. P.; Magnuson, B. A., Effects of
commercial anthocyanin-rich extracts on colonic cancer and nontumorigenic colonic
cell growth. J Agr Food Chem 2004, 52, (20), 6122-6128.
41.
Salucci, M.; Stivala, L. A.; Maiani, G.; Bugianesi, R.; Vannini, V., Flavonoids
uptake and their effect on cell cycle of human colon adenocarcinoma cells (Caco2). Brit
J Cancer 2002, 86, (10), 1645-1651.
42.
Al-Ayyoubi, S.; Gali-Muhtasib, H., Differential apoptosis by gallotannin in
human colon cancer cells with distinct p53 status. Mol Carcinog 2007, 46, (3), 176-86.
67
43.
Vermeulen, K.; Van Bockstaele, D. R.; Berneman, Z. N., Apoptosis:
mechanisms and relevance in cancer. Ann Hematol 2005, 84, (10), 627-639.
44.
Mohamad, N.; Gutierrez, A.; Nunez, M.; Cocca, C.; Martin, G.; Cricco, G.;
Medina, V.; Rivera, E.; Bergoc, R., Mitochondrial apoptotic pathways. Biocell 2005,
29, (2), 149-161.
45.
Antonsson, B., Mitochondria and the Bcl-2 family proteins in apoptosis
signaling pathways. Molecular and Cellular Biochemistry 2004, 256, (1-2), 141-155.
46.
O'Connor, L.; Strasser, A.; O'Reilly, L. A.; Hausmann, G.; Adams, J. M.; Cory,
S.; Huang, D. C. S., Bim: a novel member of the Bcl-2 family that promotes apoptosis.
Embo J 1998, 17, (2), 384-395.
47.
Miura, T.; Chiba, M.; Kasai, K.; Nozaka, H.; Nakamura, T.; Shoji, T.; Kanda,
T.; Ohtake, Y.; Sato, T., Apple procyanidins induce tumor cell apoptosis through
mitochondrial pathway activation of caspase-3. Carcinogenesis 2008, 29, (3), 585-593.
48.
Chen, W. J.; Chang, C. Y.; Lin, J. K., Induction of G1 phase arrest in MCF
human breast cancer cells by pentagalloylglucose through the down-regulation of CDK4
and CDK2 activities and up-regulation of the CDK inhibitors p27(Kip) and p21(Cip).
Biochemical Pharmacology 2003, 65, (11), 1777-1785.
49.
Kuo, P. L.; Hsu, Y. L.; Lin, T. C.; Tzeng, W. S.; Chen, Y. Y.; Lin, C. C.,
Rugosin E, an ellagitannin, inhibits MDA-MB-231 human breast cancer cell
proliferation and induces apoptosis by inhibiting nuclear factor-KB signaling pathway.
Cancer Letters 2007, 248, (2), 280-291.
50.
Liu, F.; Rothblum-Oviatt, C.; Ryan, C. E.; Piwnica-Worms, H., Overproduction
of human Myt1 kinase induces a G(2) cell cycle delay by interfering with the
intracellular trafficking of Cdc2-cyclin B1 complexes. Mol Cell Biol 1999, 19, (7),
5113-5123.
51.
Shan, B. E.; Wang, M. X.; Li, R. Q., Quercetin Inhibit Human SW480 Colon
Cancer Growth in Association with Inhibition of Cyclin D1 and Survivin Expression
through Wnt/-Catenin Signaling Pathway. Cancer Investigation 2009, 27, (6), 604-612.
52.
Hu, H. B.; Zhang, J. H.; Lee, H. J.; Kim, S. H.; Lu, J. X., Penta-O-galloyl-beta-
D-glucose induces S- and G(1)-cell cycle arrests in prostate cancer cells targeting DNA
replication and cyclin D1. Carcinogenesis 2009, 30, (5), 818-823.
68
53.
Raina, K.; Rajamanickam, S.; Deep, G.; Singh, M.; Agarwal, R.; Agarwal, C.,
Chemopreventive effects of oral gallic acid feeding on tumor growth and progression in
TRAMP mice. Molecular Cancer Therapeutics 2008, 7, (5), 1258-1267.
54.
Schumacker, P. T., Reactive oxygen species in cancer cells: Live by the sword,
die by the sword. Cancer Cell 2006, 10, (3), 175-176.
55.
Wu, X. J.; Hua, X. X., Selective killing of cancer cells by a cruciferous
vegetable derived pro-oxidant compound. Cancer Biol Ther 2007, 6, (5), 646-647.
69
ABSORPTION AND BIOLOGICAL ACTIVITIES OF POLYPHENOLS FROM
DIFFERENT MANGO (MANGIFERA INDICA L.) VARIETIES AS AFFECTED BY
Β-GLUCOSIDASE HYDROLYSIS
ABSTRACT
Biological activities of polyphenols have been related to their bioavailability.
Deglycosylation by β-glucosidases is a critical step in the absorption and metabolism of
dietary polyphenol glycosides in humans. This study aimed to evaluate the effect of the
hydrolysis of mango polyphenols with β-glucosidase on their antioxidant activity,
cancer cell-growth suppression activity and in vitro intestinal absorption through human
colon adenocarcinoma Caco-2 cell monolayers was evaluated. In addition, the
antiproliferative effect of high and low molecular weight polyphenols rich-fractions on
colon (SW-480) and breast (MDA-MB-231) cancer cells was studied. Phenolic extracts
from mango pulp contained gallic acid, mangiferin, phenolic acid derivatives and
gallotannins, which were characterized by HPLC-DAD and HPLC-ESI-MSn analysis
before and after enzymatic hydrolysis (0.17 mg β-glucosidase 1000 KU/g mango pulp/
4 h / 35°C). Phenolic acids including gallic, caffeic, ferulic, p-coumaric and phydroxybenzoic acids consisted the main compounds derived from enzymatic
hydrolysis. Caco-2 cell monolayers were incubated for 2h on the apical side with
hydrolyzed and non-hydrolyzed mango extracts. Gallic, caffeic, ferulic, p-coumaric,
vanillic and p-hydroxybenzoic acids were detected on the basolateral side for
hydrolyzed extract but only gallic acid was detected for the non-hydrolyzed extract.
High molecular weight polyphenols, mangiferin and gallotannins, were not transported.
Mango pulp polyphenols (control) from all varieties inhibited the proliferation of HT-29
colon (0-27 μg of gallic acid equiv/mL) and MDA-MB-231 breast (0-24 μg GAE/ mL)
human cancer cells by up to 99.8 and 89.9 %, respectively. Despite enhanced absorption
facilitated by enzymatic hydrolysis, a significant increase in antioxidant activity,
phenolic content and antiproliferative effects on breast and colon cancer cells was not
observed. Additionally, both high (422; 788-1852 Da) and low (138-194 Da) molecular
weight polyphenols rich-fractions equally inhibited cell proliferation of colon and breast
cancer cells at the same extent (0-20 μg of gallic acid equiv/mL), which may indicate
70
that the anti-cancer efficacy of mango polyphenolics is not dependent on enzymatic
hydrolysis.
These results corroborate previous findings from in vivo studies, which suggest
that the most of mango polyphenols are not absorbed intact through the small intestine,
but may be hydrolyzed by intestinal enzymes into low molecular weight aromatic acids,
which would be later absorbed; or when polyphenols are not absorbed, they likely reach
the large intestine, modulating the gut microflora, and thus they contribute to reduce the
risk of colon carcinogenesis.
Overall, polyphenols from several mango varieties exerted anti-cancer effects,
and these effects may not require enzymatic hydrolysis by β-glucosidase.
Keywords: Polyphenols, in vitro absorption, Caco-2 cells, cell proliferation, mango,
Mangidera indica, β-glucosidase, enzymatic hydrolysis.
71
INTRODUCTION
Mangoes (Mangifera indica L.) are one of the most important tropical fruits
marked in the world, with a global production exceeding 33 million tons in 2007.
Moreover, mangoes consist one of the most consumed fruits in Brazil due to sensorial
characteristics and nutritional value, ranking the seventh position worldwide in terms of
production in 2007 (1).
Furthermore, there has been increasing interest in understanding the mango
phytochemicals biological properties due to their health-promoting characteristics such
as antioxidant, antitumoral, anti-inflammatory, and immunomodulatory activities (2-7).
Recently, mango fruit has been listed as a nutrient-rich fruit into the unofficial
classification and so-called superfruit due to its considerable content of carotenoids,
specially β-carotene, vitamin C and polyphenols, including gallic acid, gallotannins,
quercetin and kaempferol glycosides as well as xanthone-C-glucosides (8-9).
In traditional medicine, the use of mango extracts as herbal drugs is widespread.
Vimang, a mango stem bark extract, have been used at least 10 years in Cuba medicine
with effectiveness against several diseases, like cancer (6). Moreover, studies in vitro
and in vivo performed with individual polyphenols found in mangoes have been shown
antitumor activities, such as induction of apoptosis, inhibition of tumor growth and
angiogenesis (10-15). Recently, studies performed with mango pulp extracts have been
demonstrated their chemopreventive potential (16-20), although conclusion regarding
the biological activities of the mango phenolic fraction are still not well elucidated (1718), since other bioactive compounds like L-ascorbic acid and carotenoids have been
conjunctly evaluated (16-18).
Simple phenolic compounds like phenolic acids are known to be directly
absorbed through the gut barrier by passive diffusion. However, more complex
compounds like flavonoids and procyanidins are typically transformed in the colon by
intestinal microflora into a wide array of bioactive low molecular weight metabolites,
which are later absorbed through the gut barrier (21). In humans, it has been known that
β-glucosidase activity has an important role in enhancing polyphenols absorption by
catalyzing the hydrolysis of O-linked β-glycosidic in human body (22). Therefore, the
72
health benefits from phenolic consumption are also attributed to their bioactive
metabolites and also to the modulation of the intestinal microflora (23).
Gallotannins represent the major compounds found in mango pulp, with size
ranging from 332 (mono-O-galloyl-glucose) to over 1852 Da (undeca-O-galloylglucose) or large (9). Considering that punicalagin (1084 amu) was reported to be the
highest molecular weight compound absorbed intact through the gut barrier after oral
ingestion (21, 24), it is assumed that higher molecular weight polyphenols could not be
absorbed intact in the body. However, limited bioavailability of these compounds might
not limit their biological properties, since the highest local concentration of these
compounds is found in the gut lumen, where polyphenols may act protecting colon
carcinogenesis (23, 25-27). Therefore, it is relevant to measure the biological activity of
polyphenols on cultured cells or isolated tissues in their form present in food, reflecting
the potential health benefits associated with the consumption of the entire fruit. At the
same time, it is also important to identify their metabolites and test their respective
biological activities, since the hydrolysis of glycosides and further gut bacterial
transformation of aglicones may lead to the production of more or less biologically
active compounds (23).
This study aimed to evaluate the effect of hydrolysis of mango polyphenols with
β-glucosidase on their in vitro intestinal absorption, antioxidant activity and cancer
growth suppression activity. Results from these investigations are aimed at promoting
efforts to understand the health-promoting properties of mango polyphenols.
MATERIALS AND METHODS
Plant material
Commercial varieties of mango (Mangifera indica L) Francis Kent, Ataulfo,
Tommy Atkins and Haden were kindly donated by the National Mango Board, USA.
Upon arrival at Texas A&M University, seeds and peels were removed from the fruit
and the pulp was blended, homogenized and storage at -80ºC until use.
73
Extraction of polyphenols
Frozen mango pulp (150g) was thawed and mixed with 500 mL of a 1:1:1
(v/v/v) ethanol: methanol: acetone solution, filtered with cheesecloth and whatman #1
filter paper . Solvents were evaporated at 40°C using a rotavapor (Büchi, Switzerland)
and the aqueous extract was mixed with 200mL of methanol and partionated into
hexane (1:4, v/v) in order to remove carotenoids. The hexane phase was removed and
the methanol phase centrifuged at 2000RPM for 10 min at 7 ºC to remove insoluble
precipitates. The methanol was completely evaporated using a rotavapor at 40 ºC and
polyphenols were concentrated on a C18 Waters Sep-Pak Vac 35cc 10g 20 cm3
minicolumns (Waters Corporation, Milford, MA) previously conditioned with acidified
methanol (0.01%) and nanopure water. The aqueous extract was loaded onto the C18
cartridge and washed with distilled water. Polyphenols bound to the matrix were eluted
with 100% methanol (bound fraction). The unbound fraction was partitioned three times
into ethyl acetate (EtAc) (1:4, v/v) to recover non-adsorbed polyphenols, such as
phenolic acids. Bound and unbound fraction were combined and completely evaporated
under vacuum at 40 ºC. In order to create optimal conditions to β-glucosidase activity,
polyphenols were redissolved in 0.1M citric acid buffer (pH 5.0) up to a known volume
(50mL) (Figure 12). The total phenolic content was measured spectrophotometrically by
the Folin-Ciocalteau assay (28) against an external standard of gallic acid (mg/L).
74
Crude extract (150g pulp)
(ethanol : methanol : acetone)
1:1:1 (v/v/v)
Methanol:hexane(1:4, v/v)
Aqueous extract
Evaporation
Centrifugation
Evaporation
(T <40°C)
200 X g / 10`/
7°C
(T <40°C)
C18
Unbound fraction
Pectin
Bound fraction
Elution with
ethyl acetate (3X)
Elution with
methanol (5X)
Hexane fraction
(carotenoids)
Polyphenols
(F2)
Aqueous fraction
(Sugars,
organic acids)
Polyphenols
(F1)
Evaporation
Phenolicextract
(T <40°C)
(50mL)
Figure 12. Procedure used for extraction of mango polyphenols.
Enzymatic hydrolysis
The bioavailability of dietary polyphenols is related to their absorption and
metabolism in the body (29). Although enzymes such as tannase have been extensively
used to hydrolyze tannins due to its higher efficacy in a complete degradation of the
glycoside and simultaneously release of gallic acid (8), the enzymatic hydrolysis using
β-glucosidase had the purpose of simulate the enzymatic activity in human gut, since
small intestine requires exogenous (luminal) deglycosylation by several β-glucosidases,
or cooperativity between epithelial transportes and intracellular β-glucosidases in order
to facilitate the absorption of some phenolic compounds such as flavonoid glycosides
(21, 30).
The phenolic extract (20mL, 60g pulp) was completed with 0.1M citric acid
buffer (pH 5.0) up to a known volume, which corresponded to the volume equivalent in
pulp (60mL) and incubated with β-glucosidase 1000KU ( > 2500 units/mg enzyme)
75
(MP Biomedicals, Solon, OH) at the proportion of 0.17 mg enzyme/g mango pulp for
4 h at 35° C. The same volume of extract was incubated without enzyme (control)
following the same procedures described above. In order to stop enzymatic activity,
phenolic extract was boiled for 5 min and chilled on ice immediately. Extracts were
sonicated and reeluted onto a C18 Waters Sep-Pak Vac 6cc 1g 2 cm3 minicolumns
(Waters Corporation, Milford, MA) following the same procedure described in the
previous item. The bound and unbound fractions were mixed, solvents completely
evaporated under vacuum at 40 ºC and polyphenols redissolved in 5mL of methanol
(12g pulp/mL phenolic extract). The extracts were storage at -80˚C until needed. Equal
volumes of the control and hydrolyzed extract were evaporated in a SpeedVac
concentrator (Thermo Fisher Scientific, San Jose, CA). Polyphenols (Control and
Hydrolyzed) were reconstituted with the same volume of HBSS buffer pH=6.0 for the
transepithelial transport model assay or in culture medium for cell proliferation assays.
Fractionation of mango phenolic extract
Polyphenols, the most abundant antioxidants in human diet, show considerable
differences in their structure, which is directly associated to their bioavailability,
antioxidant activity and anticancer-related properties (31-32). In order to compare the
effect of the molecular weight range of mango polyphenols on the antioxidant and
cancer cell growth suppression activities, Ataulfo hydrolyzed extract (5 mL) was
fractionated into low and high molecular weight polyphenols-enriched fractions, as
described in Figure 13. Polyphenols were bound onto a C18 Waters Sep-Pak Vac 6cc 1g
2 cm3 minicolumns (Waters Corporation, Milford, MA), previously conditioned with
acidified methanol and water, and sequentially eluted with methanol 25% (v/v) and
methanol 100%(v/v). The methanolic fraction (F1) was evaporated and the volume was
made to 5 mL with methanol. The unbound fraction was partitioned three times into
ethyl acetate (EtAc) (1:4, v/v). The MeOH 25% fraction was evaporated at < 40°C and
the aqueous extract was reeluted again on C18 minicolumns. The polyphenols were
washed with 100% methanol and mixed with the ethyl acetate fraction. The solvents
were completely evaporated and the volume made to 5 mL with methanol (F2). F1
consisted mainly of high molecular weight (HMW) polyphenols, whereas F2 was
76
enriched with polyphenols within a lower molecular weight range (LMW). The extracts
were storage at -80˚C until needed. Equal volumes of F1 and F2 fractions were
evaporated in a SpeedVac concentrator (Thermo Fisher Scientific, San Jose, CA).
Polyphenols were reconstituted in culture medium for cell proliferation assays.
Phenolic extract
(MeOH)
Evaporation
(T < 40°C)
C18
Bound fraction
Elution with
25% MeOH (1X)
Unbound fraction
LMW polyphenolics
(Phenolic acids)
Elution with
ethyl acetate (3X)
Evaporation
(T < 40°C)
Elution with
100% MeOH (1X)
Aqueous fraction
(Sugars,
organic acids)
Elution with
100% MeOH (1X)
HMW polyphenolics (F1)
(Gallotannins + mangiferin)
LMW polyphenolics
Evaporation
Phenolic acids
(T < 40°C)
LMW polyphenolics (F2)
(Phenolic acids)
Figure 13. Procedure used for fractionation of mango phenolic extract.
HPLC-DAD and HPLC-ESI/MSn Analysis
Polyphenols were analyzed using a Waters Alliance 2690 (Milford, MA, USA)
HPLC system equipped with a Symmetry ® C18 5 µm column, 4.6 mm x 250 mm
column and a 4.6 mm x 20 mm guard column. Individual phenolic compounds were
separated with mobile phases of nanopure water: acetic acid (98:2, v/v) (phase A) and
nanopure water:acetonitrile:acetic acid (68:30:2, v/v/v) (phase B) at a flow rate of 0.8
77
mL/min with PDA detection from 210 to 600 nm. Phenolic compounds were separated
with a gradient program that ran phase A from 100-70% in 20 min; 70-50% in 10 min;
50-30% in 20 min; and 30-0% in 20 min, 100% phase B in 15min, and returning to the
original composition in 3 min for column equilibration. Monitoring was performed at
250 nm (p-hydroxybenzoic acid and derivatives), 280nm (gallotannins) and 340nm
(hydroxycinnamic acids, flavonols and xanthones).
Additional identification by mass spectrometric analyses was performed on a
Thermo Finnigan LCQ Deca XP Max MSn ion trap mass spectrometer equipped with
an ESI ion source (Thermo Fisher, San Jose, CA). Polyphenolic extracts were analyzed
using a Waters Alliance 2690 (Milford, MA, USA) HPLC system equipped with a
Dionex Acclaim ® 120 C18 5 µm column, 4.6 mm x 250 mm column and a 4.6 mm x
20 mm guard column. Phenolic compounds were separated using 0.5% formic acid in
water (solvent A) and 0.5% formic acid in acetonitrile (solvent B) as mobile phases, at a
flow rate of 0.4 mL/min. Gradient program ran phase A for 5 min, from 100-70% in 25
min; 70-50% in 15 min; 50-0% in 25 min; and returning to original composition in 8
min.
Ionization was conducted in the negative ion mode under the following
conditions: sheath gas (N2), 40 units/min; auxiliary gas (N2), 5 units/min; spray
voltage, 5 kV; capillary temperature, 350 °C; tube lens offset, 10 V.
Identification of phenolic compounds was performed by their comparison of
retention times and UV-visible spectral characteristics with those obtained from
standards (Sigma-Aldrich, St Louis, MO) and mass spectrometric properties.
Antioxidant capacity (ORAC assay)
Hydrophilic antioxidant capacity of mango phenolic extracts, excluding further
contribution of L-ascorbic acid, reducing sugars, and soluble proteins, was measured
using the oxygen radical absorbance capacity (ORAC) assay, as described by Talcott
and Lee (2002) (33). Peroxyl radicals were generated by 2,2‘-azobis(2-amidinopropane)
dihydrochloride, and fluorescence loss was monitored on a FLUOstar fluorescent
microplate reader (BMG Labtech Inc, Durhan, NC) at 485 nm excitation and 538 nm
emission. Phenolic extracts were equally diluted in pH 7.2 phosphate buffer in a 50, 100
78
and 200-fold. Data were expressed in μmol Trolox equivalent/100g mango pulp or μmol
Trolox equiv/L phenolic extract.
Cell culture
Cell lines were obtained from the American Type Culture Collection (ATCC,
Manassas, VA) and cultured according ATCC recommendations. HT-29 human colon
adenocarcinoma cells were cultured using McCoy`s 5A modified medium with 1.5 mM
L-glutamine (Gibco™, Invitrogen Corp., Grand Island, NY). The estrogen independent
MDA-MB-231 breast cancer cells were cultured using Dulbecco’s modified Eagle’s
medium (DMEM) high glucose, with 2 mmol/l L-glutamine, without sodium pyruvate
and with phenol red. Culture mediua were supplied by Invitrogen (Gibco™, Invitrogen
Corp., Grand Island, NY). Both media were supplemented with 10% (v/v) fetal bovine
serum (FBS) and 1% Penicillin-Streptomycin antibiotic mix. Cells maintained at 37 ºC
with a humidified 5% CO2 atmosphere for propagation. A 2.5% FBS was used in
culture medium for cell proliferation experiments. Cells were seeded overnight (1.5 x
104 cells/well) in a 24 well plates and treated with the same dilutions of hydrolyzed and
control phenolic extracts from the varieties Haden, Francine, Kent, Ataulfo and Tommy
Atkins, incubated with and without β-glucosidase.
Cell proliferation
Total cell numbers were indicative of the cell growth of cancer cells and the
antiproliferative effects of mango polyphenols were determined following 48h of
incubation, using a Beckman Coulter Particle Counter (Fullerton, CA). The difference
in cell-number between final incubation time (48h) and the incubation start (0h)
represents net growth. The polyphenols ability to inhibit the cell growth by 50% was
expressed in IC50, as mg gallic acid equivalent/L phenolic extract.
79
Transepithelial Transport Model
Caco-2 colon carcinoma cells were obtained from American Type Culture
Collection (ATCC, Manassas, VA) and cultured at 37°C and 5% CO2 in Dulbecco’s
modified Eagle’s medium (1X) high glucose (DMEM) containing 20% fetal bovine
serum (FBS), 1% non-essential amino acids, 100 units/mL penicillin G, 100 µg/mL
streptomycin, 1.25 µg/mL amphotericin B, and 10 mM sodium pyruvate (chemicals
supplied by Sigma-Aldrich Co., St. Louis, MO). Cells were seeded (100,000
cells/insert) onto a Costar Transwell insert, 0.4 μm pore diameter, 12 mm transparent
polyester cell culture insert well plates (Transwell, Corning Costar Corp., Cambridge,
MA), with 0.5 mL of DMEM 15% FBS in the apical side and 1.5 mL in the basolateral
side. Fresh medium was replaced every 2 days. Cells (passages 5-15) were grown and
differentiated to confluent monolayerss for 21 days (34). Cells confluence and
monolayers integrity were evaluated by the transepithelial electrical resistance values
(TEER), measured with an EndOhm Volt ohmmeter equipped with a STX-2 electrode
(World Precision Instruments Inc., Sarasota, FL) after 30 min of incubation with HBSS
and 2h of incubation with the treatments. Monolayerss with TEER values >450 Ω cm2
at the beginning and >225 Ω cm2 at the end were used for transport experiments.
Additional control of the monolayers integrity was performed using lucifer yellow, a
very hydrophilic probe which can permeate the cells only trough the paracellular route.
This probe (100 μg/ml) was added to the apical side, incubated for 1h and the % of
passage was determined by comparing with fluorescence obtained from a standard
solution using excitation and emission wavelengths 485 and 530 nm, respectively.
Prior transport experiments, cells were rinsed with Dulbecco’s phosphate buffer
saline (DPBS) and growth media was replaced by Hank’s balanced salt solution (HBSS,
Fischer Scientific, Pittsburgh, PA) containing 10 mM 2-(N-morpholino)ethanesulfonic
acid solution (MES, Gibco BRL Life Technology, Grand Island, NY) adjusted to pH 6.0
in the apical side, and HBSS containing N-[2-hydroxyethyl]piperazine-N’-[2ethanesulfonic acid] buffer solution (1 M) (HEPES, Gibco BRL Life Technology,
Grand Island, NY) adjusted to pH 7.4 in the basolateral side, creating a pH gradient
similar to the small intestine environment. Mango polyphenols (control and hydrolyzed
phenolic extracts) were equally diluted in HBSS adjusted to pH 6.0 (
80
3,000 mg
gallic acid equivalent/L) to be used in cell culture. After incubation at 37°C for 30min
of Caco-2 cells, the HBSS adjusted to pH 6.0 from apical side of cell monolayerss, was
replaced by 500 µL of phenolic extracts. Sample aliquots (200 µL) were taken from the
basolateral compartment at 30, 60, 90, and 120 min, and immediately acidified with 5μL
of 4N HCl, kept frozen (-80°C), and analyzed within one week. Samples were filtered
through 0.45µm PTFE membranes (Whatman, Florham Park, NJ) and injected directly
into the HPLC-PDA system.
Statistical Analysis
Quantitative data represent means with the respective standard deviation (SD) of
three or more replicates. Statistical analysis was performed using JMP 5 statistical
software (SAS Institute, 2002). One-way analysis of variance (ANOVA) and TukeyKramer HSD test (p<0.05) were used to compare the effect of different varieties on the
analyzed variables and paired t-student to compare the effect of polyphenols hydrolysis
on cancer cell growth suppression, total phenolic content and antioxidant activity.
Pearson correlation coefficient was estimated to determine the correlation between
phenolic content and antioxidant capacity.
RESULTS AND DISCUSSION
Mango pulp phenolic content and antioxidant activity
The total phenolic content (TPC), analysed by Folin-Ciocalteau assay, from the
mango varieties Ataulfo (At), Haden (Ha), Kent (Ke), Francis (Fr) and Tommy Atkins
(To) ranged from 15.1 to 55.5 mg GAE/100g pulp, according to Table 5, and At
contained the highest TPC. Previous studies showed that mango TPC may range from
48.4 to 294 mg GAE/ 100g (35-37), as influenced by variety, ripening stage, geographic
production, harvesting, processing and storage conditions. These factors might affect
the mango phytochemical composition, and consequently, their antioxidant capacity (8,
37). Thus, determination of total polyphenols may be relevant when analyzing the
antioxidant and anticarcinogenic activities from fruit, since both are in part attributable
81
to the polyphenols content. Our results showed that the antioxidant activity (AA) of
mango pulp phenolic extract, determined by ORAC assay (33), correlated to the TPC
(r = 0.96), and At polyphenols showed to be the most effective in protecting the
fluorescent molecule from the peroxyl radicals-induced oxidative degeneration (288.7 ±
10.2 µmol Trolox equivalent/100g pulp respectively) (Table 5). These results are in
accordance with previous findings reported by Kim and co-works (2007) (36). They
showed that the decrease of mango AA positively correlated (r = 0.98) to the reduction
of the TPC during ripening, even though the presence of other compounds in the mango
clarified juice such as L-ascorbic acid, reducing sugars and soluble proteins have
contributed to increase the ORAC value (36, 38). Therefore, in addition to the influence
of ripening stage, variety and other factors as previously described in literature, the
contribution of non-phenolic compounds for the AA of mango pulp extracts might also
explain the higher ORAC values reported in literature, ranging from 326 to 1002 μmol
TE/100g pulp (36, 38).
Table 5. Total phenolic content and antioxidant activity of polyphenols extracted from
different mango varieties.
Mango
Variety
Ataulfo
Phenolic content
(mg GAE/L)
1631.0 ± 48.4 a
Phenolic content
(mg GAE/100g pulp)
55.5 ± 1.6 a
Antioxidant activity (µmol
TE/100g pulp)
288.7 ± 10.2a
Francis
577.3 ± 7.7 b
19.2 ± 0.3 b
198.2 ± 14.4b
Tommy
454.0 ± 28.6 c
15.1 ± 0.9 c
180.8 ± 12.6bc
Haden
498.1 ± 3.4 c
16.6 ± 0.1 c
164.0 ± 17.2c
17.8 ± 0.0 bc
160.3 ± 4.0c
Kent
547.6 ± 22.3 bc
Data are means of three or more independent determinations ± SD. Means with different letters in the
column are statistically different at p< 0.05 (Tukey-Kramer HSD test).
Enzymatic hydrolysis of mango pulp polyphenols
Phenolic content and antioxidant activity
The enzymatic hydrolysis of polyphenols extracted from the varieties Fr, Ha and
To, following 4h of incubation with β-glucosidase at 35 °C, did not statistically affect
the TPC and the AA from their phenolic extracts (Table 6). On the other hand, the
hydrolyzed extracts of the varieties At and Ke showed a 28 % and 34.2 % lower than
82
those from control extracts, respectively (Table 6). This reduction may be associated to
the formation of a precipitate, which could be visually observed following 4h of mango
polyphenols incubation with β-glucosidase and was latter removed following elution of
polyphenols onto C18 mini-column. However, even though the precipitation formation
may occur likely related to tannin-protein interaction, in biological systems the same
phenomena may not occur, since the stoichiometry of interacting species would tend to
lead to soluble complexes only (39).
Table 6. Total phenolic content and antioxidant activity of hydrolyzed and control
mango phenolic extracts.
Cultivar
Phenolic content
(mg GAE/L)
Hydrolysed
Control
a
4102.0 ± 8.2 b
2953.9 ± 0.7
Ataulfo
Tommy
1344.7 ± 7.6 a
1345.8 ± 8.5 a
Atkins
1655.9 ± 15.4 a
1732.1 ± 27.7 a
Francis
a
1814.6 ± 4.3 b
1193.6 ± 69.8
Kent
1205.7 ± 14.4 a
1159.8 ± 51.9 a
Haden
Data are means of three or more replicates ± SD. Means with
different at p< 0.05 (paired t-student test).
Antioxidant activity
(µmol TE/L)
Hydrolysed
Control
a
15365.6 ± 4.0
23337.5 ± 42.7 b
16113.5 ± 101.3 a
17980.0 ± 286.0 a
10749.6 ± 429.0 a
11457.9 ± 412.4 a
different letters in the
15870.4 ± 92.1 a
17093.7 ± 158.9 a
15341.4 ± 38.5 b
11309.4 ± 142.6 a
line are statistically
Hydrolyzable tannins consist of a polyol (typically β-D-glucose) partially or
totally esterified with phenolic groups such as gallic acid (gallotannins) or ellagic acid
(ellagitannins) (40-41). In gallotannins, galloyl groups are hydrophobic sites which can
interact with aliphatic side chains of amino acids through hydrophobic association to
form complexes (42). Interactions of tannins and proteins are essentially a dynamic
surface phenomenon, generally reversible, which involves most frequently hydrophobic
effects, which are reinforced by hydrogen bonds. Gallotannins bound more to proteins
(histone, bovine serum albumin, casein and gelatin) and phospholipids (L-a-lecithin, La-cephalin and sphingomyelin) than to sugars (42). Relative affinities of tannins for
different proteins may vary as much as 10000-fold (43-44). Tannins have lower affinity
for compact globular proteins and preferential binding to proline-rich proteins, possibly
due to more open and flexible conformations, which provides better access to
polyphenol-binding sites (43, 45). Therefore, the polypeptide chain characteristics of
some β-glucosidases, which contain highly conserved peptide motifs like Thr-Phe-Asn-
83
Glu-Pro (TFNEP) and Ile-Thr-Glu-Asn-Gly (ITENG) (46-48), suggest favorable tannininteraction affinities. Binding affinities to protein, phospholipids and sugars are, in part,
function of molecular size of gallotannins, which is enhanced by the addition of each
galloyl group and reaches a maximum in the flexible disk-like structure of penta-Ogalloyl-D-glucose (42). These evidences led to the inference that high molecular weight
gallotannins should precipitate proteins more effectively. However, some studies
suggest that this rule is an oversimplification and does not apply to all tannins because
the biochemical properties of gallotannins isomers with the same molecular weight,
including susceptibility to hydrolysis as well as ability to precipitate proteins, are
structure-dependent (49-50). Studies have been shown that conformational mobility and
flexibility of either the tannin or the protein are as significant as molecular size.
Conformational freedom is necessary for strong binding, and therefore, gallotannins
should precipitate certain proteins more efficiently than ellagitannins, which are
conformationally rigid. Based on these evidences, the presence of gallotannins in At and
Ke might explain the reduction on TPC after hydrolysis as a result of protein-tannin
interaction-derived complex, although no significant reduction in TPC from the other
varieties was found.
Likewise, the antioxidant capacity of Fr, Ha and To was not significantly
affected by enzymatic hydrolysis (Table 6). In contrast, as a consequence of the
reduction in the amount of hydroxyl groups present in the phenolic extract likely
associated to tannin-protein interaction, a significant decrease ( p < 0.05) in the
antioxidant activity of At and Ke hydrolyzed phenolic extracts was observed when
compared to AA from control extracts (Table 6). These results suggest considerable
contribution of hydrolyzable tannins to antioxidant capacity of mango pulp.
HPLC-DAD and HPLC-ESI-MSn analysis
Mango pulp polyphenols from phenolic extracts (
1000 ppm) from all
varieties were characterized by HPLC-DAD and HPLC-ESI-MSn analysis. Phenolic
profile from control extracts varied depending on the variety (Figure 14). Phenolic acids
and the xanthone mangiferin were characterized unambiguously following HPLC
analysis. Mango pulp consisted of free forms and derivatives of hydroxycinnamic acids
84
caffeic, ferulic and p-coumaric acids, as well as free and esterified forms of gallic acid
and p-OH-benzoic acid. Gallic and ferulic acids were detected in all varieties; caffeic
acid was found in At, Ha and Ke; p-coumaric acid was detected in At, To, Fr; and Ke
and p-OH-benzoic acid just detected in Ha. Mangiferin was identified in the most of the
varieties, but not in Francis (Table 7). Gallotannins, which show UV spectral
similarities to gallic acid at 280 nm comprised the predominant polyphenols in mango
pulp, which chemical structure varied from one (mono-O-galloyl-glucoside) to eleven
(undeca-O-galloyl-glucoside) units of gallic acid esterified in a glucose core, as
characterized by HPLC-ESI-MSn analysis (Table 7, Apendice). Similarly, gallotannins
with 4-7 galloyl groups surrounding a glucose core were previously identified in mango
pulp from the variety Tommy Atkins (9). Gallotannins with molecular weight higher
than 939 showed elution pattern after 40 min (Figure 14). Even though flavonol and
xanthone glycosides were hypothesized by monitoring at 340 nm, flavonol-3-Oglycosides, represented by quercetin and kaempferol glycosides, as well as xanthone-Cglucosides, as previously found in mango pulp (8-9), were not identified in our extracts.
3
3
(A)
0.60
AU
3
0.40
0.20
3
3
3
6
1
33
33
3
0.00
10.00
20.00
30.00
40.00
50.00
Minutes
60.00
70.00
80.00
AU
0.10
0.05
(B)
6
8
5
0.00
0.00
10.00
20.00
30.00
7
40.00
50.00
Minutes
60.00
70.00
80.00
Figure 14. Representative chromatograms at 280 nm (A) and 340 nm (B) of Ataulfo
mango pulp polyphenols from control phenolic extracts. Peak assignments: (1) gallic
acid; (2) p-OH-benzoic acid; (3) gallotannins ; (4) vanillic acid; (5) caffeic acid; (6)
mangiferin; (7) p-coumaric acid; (8) ferulic acid.
85
Table 7. HPLC-DAD and HPLC-ESI-MSn of mango phenolic extracts (control) from
the varieties Ataulfo, kent, Francis, Tommy Atkins and Haden.
Class
Phenolic acids
Xanthones
Polyphenols
Gallic acid
p-coumaric-acid
Ferulic acid
Caffeic acid
p-OH-benzoic acid
Vanillic acid
mangiferin
Ataulfo
Haden
Kent
Francis
9
9
9
9
nd
nd
9
9
nd
9
9
9
nd
9
9
9
9
9
nd
nd
9
9
9
9
nd
nd
nd
nd
9
9
9
9
mono-O-galloyl- glucose
Gallotannins
di-O-galloyl- glucose
nd
9
tetra-O-galloyl- glucose
nd
9
9
9
nd
nd
Tommy
Atkins
9
9
9
nd
nd
nd
9
9
nd
nd
penta-O-galloyl- glucose
9
9
9
9
9
hexa-O-galloyl- glucose
9
9
9
9
9
hepta-O-galloyl- glucose
9
9
9
9
9
octa-O-galloyl- glucose
9
9
9
9
9
nona-O-galloyl- glucose
9
9
9
9
9
deca-O-galloyl- glucose
9
nd
nd
nd
nd
undeca-O-galloyl-glucose
9
nd
nd
nd
nd
nd= not identified.
Most of dietary polyphenols are found in the form of esters, glycosides, or
polymers that cannot be directly absorbed in their intact form through small intestine.
Thus, deglycosylation of polyphenols by several β-glucosidases are known to facilitate
the absorption of some phenolic compounds as aglicones (21, 30). The human cytosolic
β-glucosidase (hCBG), a group of nine enzymes with similarities in the amino-acid and
structural features related to their substrate specificities, are present in the cytosol of
liver, spleen, kidney, small intestine and lymphocytes of mammals (51), and βglucosidase activity has been related to the intestinal absorption of glycosides in vivo
and through Caco-2 cell monolayers (52-57).
As a result of in vitro enzymatic hydrolysis by β-glucosidase, changes in the
chemical structure and concentration of individual polyphenols were observed, mainly
regarding the profile of phenolic acids derivatives. On the other hand, no significant
structural changes were observed in gallotannins, which remains as the major
polyphenols in hydrolyzed extracts (Figure 15).
86
0.80
3
1
3
(A)
0.60
AU
3
0.40
3
2
0.20
3
3
8
6
4
5
7
33
0.00
10.00
20.00
30.00
0.16
0.14
40.00
50.00
8
Minutes
60.00
70.00
80.00
(B)
0.12
AU
0.10
6
0.08
0.06
5
0.04
7
0.02
0.00
0.00
10.00
20.00
30.00
40.00
Minutes
50.00
60.00
70.00
80.00
Figure 15. Representative chromatograms at 280 nm (A) and 340 nm (B) of Ataulfo
mango pulp polyphenols from hydrolyzed phenolic extracts. Peak assignments: (1)
gallic acid; (2) p-OH-benzoic acid; (3) gallotannin; (4) vanillic acid; (5) caffeic acid; (6)
mangiferin; (7) p-coumaric acid; (8) ferulic acid.
Caffeic and p-coumaric acids, not previously detected in control extracts from Fr
and Ha, respectively, were identified after hydrolysis. Indeed, derivatives of cinnamic
acids, mainly represented by caffeic, ferulic, sinapic and p-coumaric acids, are rarely
found in free form, but in general, they are esterified with quinic, tartaric acids or
carbohydrates derivatives (29). Also, p-hydroxybenzoic acid was detected after
hydrolysis in At and Fr extracts. Hydroxybenzoid acids are commonly found in a
limited distribution in foods and have as the main representatives gallic, ellagic,
protocatechuic and 4-hydrobenzoic acids (29, 58).
Vanilic acid, not previously identified in mango pulp, was identified in
hydrolyzed extracts from At and Fr. This phenolic acid is a catabolic product of ferulic
acid degradation, which was likely converted due to enzymatic treatment conditions like
pH, or by enzyme, since some β-glycosidases have the ability to convert ferulic acid to
87
vallinic acid (58). Therefore, it is not hypothesized the presence of this compound in
mango pulp.
Despite the considerable source of gallic acid due to the predominance of
gallotannins with high level of galloylation in mango pulp, little increase in its
concentration was observed (Table 8), suggesting that β-glucosidases was not able to
completely hydrolyze gallotannins present in mango pulp. It might be related to the
higher amount of gallic acid esterified in the glucose moiety or linked with another
moiety of gallic acid via depsidic bond. Indeed, in contrast to tannases, which can
hydrolyze ester and depsidic bonds between two galloyl residues in hydrolysable
tannins (59-61), β-glucosidases catalyze only the hydrolysis of O-linked β-glycosidic
bonds at the non-reducing end of carbohydrates with retention of anomeric
configuration (51). Although these enzymes are capable of deglycosylate a broad
variety of glycosides including aryl- and alkyl-β-D-glycosides, their physiological
function may varies greatly depending upon their origin (plants, fungi, animals or
bacteria) and substrate specificity (22). Regarding flavonoids, for instance, the human
cytosolic β-glucosidase shows high specificity for 4- and 7-glucosides of isoflavones,
flavonols, flavones and flavanones, but does not hydrolyze 3-linked flavonoids
glucosides (22). However, the mechanisms related to the hydrolysis of hydrolyzable
tannins by β-glucosidases are still unclear.
Table 8. Effect of enzymatic hydrolysis on the phenolic acids profile of mango
phenolic extract.
Increase (fold) in polyphenols area after hydrolysis
Tommy
Class
Polyphenols
Ataulfo
Haden
Kent
Francis
Atkins
Gallic acid
14.1
1.0
7.2
10.4
2.4
p-hydroxybenzoic acid
ah
17.4
nd
nd
ah
Phenolic
Vanillic acid
ah
Nd
nd
nd
ah
acids
Caffeic acid
19.6
7.2
16.7
nd
ah
p-coumaric-acid
24.0
Ah
32.1
11.6
40.8
Ferulic acid
123.2
79.9
1159.3
89.0
38.8
nd= not identified, ah = phenolic acids detected after enzymatic hydrolysis
Overall, the concentration of phenolic acids in all varieties, analysed by HPLC,
following 4h of incubation with β-glucosidase, increased from around 2.4 (gallic acid)
to 1159.3-fold (ferulic acid), which were the main phenolic compounds resulted from
88
β-glucosidase activity (Table 8). These results corroborate in vivo findings regarding the
availability and metabolism of polyphenols in humans, which show that β-glycosidases
activity in human gut may hydrolyze glycosides into aglicones with low molecular
weight, including phenolic acids(62-63).
Transepithelial Transport Model
Transport of mango polyphenols, hydrolyzed or not, was studied using Caco-2
cell monolayerss from the apical to the basolateral side. Caco-2 human colon cells are
widely accepted as an in vitro system for predicting the bioavailability of drugs and
others compounds and has been extensively used to predict the in vivo intestinal
absorption of several polyphenols, including phenolic acids (64), flavonoids (65), and
procyanidins (66). However, absorption studies using Caco-2 cells have been shown
some limitations, which may not correctly predict the metabolism of polyphenols in
human, since the deglycosylation step may not be involved (30). Nemeth et al. (2003)
(30) showed lack or low level of β-glucosidase activity towards lactose and quercetin
glucosides in fully differentiated Caco-2 cells incubated over 27 days, whereas high
activity was observed for β-glucosidases (lactasephlorizin hydrolase and cytosolic β –
glucosidase) isolated from samples of human small intestine and liver, leading to the
conclusion of low activity of cytosolic β-glucosidase in Caco-2 cells. Therefore,
previous hydrolysis of mango polyphenols shows to be a critical step to correctly
predict their absorption in vivo. Hydrolyzed and control phenolic extracts at the same
dilution (TPC
3000 mg GAE/L) were loaded into the apical side of the cell
monolayerss and individual polyphenols concentrations were analyzed over time by
HPLC. Analytical HPLC chromatograms of mango polyphenols transported in the
apical- basolateral direction through Caco-2 cell monolayers following 2h of incubation
are present in Figure 15.
Phenolic acids were the mango polyphenols better transported through Caco-2
cells monolayers (Figure 16), whereas no absorption evidence was found to high
molecular weigh compounds comprising gallotannins and mangiferin. Ferulic (FA) and
p-coumaric acid (PC) were the most absorbed in cells treated with hydrolyzed phenolic
extract, following p-OH-benzoic acid, caffeic (CA) and gallic acid (GA) (Table 9).
89
0.006
(A)
0.002
1 8 .5 3 7
AU
0.004
0.000
-0.002
0.00
10.00
20.00
30.00
40.00
Minutes
0.004
50.00
60.00
70.00
80.00
60.00
70.00
80.00
60.00
70.00
80.00
(B)
AU
0.002
0.000
1
-0.002
10.00
0.015
(C)
(H)
30.00
40.00
50.00
6
Minutes
2
AU
0.010
20.00
3
0.005
5
1
4
0.000
0.00
10.00
20.00
30.00
40.00
Minutes
50.00
Figure 16. Chromatograms (280 nm) from samples taken from the basolateral
compartment of Caco-2 cells following 2h of incubation with HBSS (pH 6.0) (A),
control (B) and hydrolyzed (C) Ataulfo phenolic extracts. Peak assignments: (1) gallic
acid; (2) p-OH-benzoic acid; (3) vanillic acid; (4) caffeic acid; (5) p-coumaric acid; (6)
ferulic acid.
90
Table 9. Absorption (%) of phenolic acids through Caco-2 colon cancer cells following
2h incubation with control and hydrolyzed extracts.
Polyphenols
Gallic acid
Control
Concentration
Absorption
(mg GAE/L)
(%)
8.8
12.7 ± 0.6
Hydrolyzed extract
Concentration
Absorption
(mg GAE/L)
(%)
66.6
2.3 ± 0.2
p-OH-benzoic acid
nd
-
61.7
48.8 ± 4.4
Vanillic acid
nd
-
8.8
134.2 ± 13.6
Caffeic acid
nd
-
7.2
13.7 ± 1.4
p-coumaric-acid
nd
-
9.3
70.7 ± 7.5
nd
-
29.5
74.5 ± 8.8
Ferulic acid
nd= not detected
Our findings are in agreement with previous in vivo and in vitro studies using a
Caco-2 cell monolayers (64-67) and evidence the influence of molecular weight and
chemical structure on bioavailability of polyphenols. It has been demonstrated that GA,
as well as m-hydroxybenzoic acid, are transported across human intestinal Caco-2 cells
via a paracellular route, which permeability is characterized as a non-polarized, dosedependent and pH independent (64, 68-70), whereas FA and PC are absorbed in Caco-2
cells by monocarboxylic acid transporter (MCT) (64, 68). CA, on the other hand, is
absorbed by MCT and mainly by paracellular diffusion, in a non-saturable transport
(69). As a result of different absorption mechanisms, studies have demonstrated higher
in vivo absorption efficiency of PC as compared to GA, which showed permeation rates
70-100 fold lower than that of PC, when administrated in rats at the same doses (64,
71). Even though GA (61.7 mg GAE/L) was present in higher concentration in mango
hydrolyzed phenolic extract, its absorption was the lowest detected when compared to
the other phenolic acids permeability. Interestingly, despite GA concentration in control
extract was 7.5- fold lower than in hydrolyzed extracts, gallic acid was 5.5 times more
absorbed in control extracts-treated cells (table 9). In contrast to previous in vivo
studies, that found similar concentrations of GA in urine of rats after consumption of the
same amount of GA as a pure compound and in black tea (72-73), our results suggested
that the presence of other phenolic acids might significantly influence the GA
availability (Table 9). Even though some studies have been demonstrated the influence
of food matrix on polyphenols availability (74-75), specific studies with GA taking into
91
account this issue have not been afforded yet. For instance, Konishi et al. (2004) (71)
suggested that p-coumaric acid in vivo absorption was inhibited by benzoic acid, likely
by inhibition of both MCT-mediated and the passive diffusion transport of PC.
It has been shown that the bioavailability of polyphenols is dependent to their
physiochemical properties (27). For instance, the glycosylation influences chemical,
physical and biological properties of a polyphenol and directly affect its polarity and
partition coefficient, which is thought to govern the intestinal absorption of dietary
polyphenols, because only passive diffusion seems to be involved (27). According to the
Lipinski`s Rule of 5, compounds with five or more hydrogen bond donors (OH and NH
groups), ten or more hydrogen bond acceptors (N and O), molecular weight higher than
500, and Log P greater than 5 are usually poorly absorbed following oral administration
(76-77). Indeed, several in vivo studies support this rule (71, 75, 78). In general, when
hydroxycinnamic or hydroxybenzoic acids were ingested under free form, they were
rapidly absorbed in vivo from the stomach or the small intestine and they were
conjugated by the intestinal and/or hepatic detoxification enzymes. Studies carried by
Konish et al. (2004) (71) in rats suggested rapid absorption of GA in the upper part of
the gut, from the stomach, from small intestine, or both. Investigations on
hydroxycinnamic acids shown that when ingested under free form, they are rapidly
absorbed from the stomach and small intestine. In rats, ferulic acid (FA) is well
absorbed in the upper part of the rat gut (75, 79-80), whereas in humans, it has been
rapidly detected in the plasma with urinary excretion reaching its maximum in 7 or 8 h
after food ingestion (81-84). Similarly to FA, CA is well absorbed, and whatever the
administrated dose in animals, most of the different metabolites disappear after 6h (8586). Likewise, PC is also rapidly absorbed in the gastrointestinal tract when orally
administrated in rats in its intact form (71). In addition, phenolic acids like FA, PC, CA
and GA are better absorbed when administrated in humans and rats in free form as
compared to esterified form (29).
A commonly accepted concept regarding polyphenols intestinal absorption is
that glycosylated polyphenols need to be converted to the aglycone by glucosidases in
the food or gastrointestinal mucosa, or from the colon microflora in order to be
absorbed by passive diffusion (87). Overall, gallotannins upon hydrolysis yield glucose
and gallic acid, whereas ellagitannins undergo lactonization to produce ellagic acid (23).
92
Punicalagin (PU), a water soluble ellagitannin found in pomegranates with molecular
weight of 1084 (88), is known to be the highest molecular weight compound ever
absorbed through the gut barrier after oral ingestion (21, 24), which was hydrolyzed into
ellagic acid in vivo(89), and in vitro across the mitochondrial membrane (90).
Some studies have been shown bioconversion of ferulic acid into vanillic acid by
β-glycosidases (58, 91-92), which may explain the absorption of vanillic acid over
100% (Table 9). This data suggest beta-glucosidase activity through intestinal
monolayers.
Overall, our findings suggest that β-glucosidases may hydrolyze mango
polyphenols in vivo into low molecular weight compounds, mainly phenolic acids,
which are later absorbed through gut barrier. On the other hand, β-glucosidases activity
did not contribute to enhance absorption of high molecular weight polyphenols, such as
gallotannins. Because these polyphenols are not likely absorbed in vivo and reach the
large intestine, they may modulate gut microflora and act protecting colon
carcinogenesis.
Cell-growth supressive activity of mango pulp polyphenols
Mechanisms associated to the chemopreventive effects of polyphenols comprise
inhibition of carcinogen activation, induction of phase II enzymes, inhibition of
arachidonic acid metabolism, modulation of different oncogenes, tumor suppressor
genes, and signal transduction pathways, leading to cell proliferation inhibition,
transformation, angiogenesis as well as induction of apoptosis (93).
Despite the biological activities of polyphenols has been related to their
availability, studies indicate that health effects of polyphenols may not require their
absorption through the gut barrier, since the highest local concentration of these
compounds is found in the gut lumen (94). Therefore, it is relevant to measure the
biological activity of polyphenols on cultured cells or isolated tissues in their form
present in food. At the same time, polyphenols are extensively metabolized both in
tissues and by the colonic microflora into a wide range of low molecular weight
compounds, which may have lower or higher biological activities. Thus, it is important
to identify their metabolites and test their respective biological activities. In this context,
93
cell-growth suppressive effects of mango polyphenols and their metabolites derived
from hydrolysis with beta-glucosidase were studied using cancer cell culture models.
The antiproliferative activities of control and hydrolyzed mango phenolic
extracts were evaluated in a cell culture model using MDA-MB-231 breast and HT-29
colon cancer cells. Polyphenols extracted from the pulp of all varieties induced a
concentration-dependent cell proliferation inhibition on human HT-29 colon and
estrogen independent MDA-MB-231 breast human cancer cells (Figure 17). Mango
pulp polyphenols inhibited colon (0-27 μg of gallic acid equiv (GAE)/mL) and breast
(0-24 μg GAE/mL) cancer cell proliferation by up to 99.8 and 89.9 %, respectively.
(A.c)
100
Ataulfo
Kent
Francis
Tommy Atkins
Haden
100
80
60
40
20
80
60
40
20
0
0
10
20
30
(A.h)
100
0
40
Polyphenolics (mg GAE/L) Kent
Ataulfo
Tommy Atkins
Haden
Francis
10
20
Polyphenolics (mg GAE/L)
120
(B.h)
100
Net Growth of HT-29 colon
cancer cells (% of control)
0
120
Net Growth of MDA-MB-221 breast
cancer cells (% of control)
(B.c)
120
Haden
Francis
Tommy Atkins
Kent
Ataulfo
Net Growth of HT-29 colon
cancer cells (% of control)
Net Growth of MDA-MB-231 breast
cancer cells (% of control)
120
80
60
40
20
30
40
Haden
Kent
Francis
Ataulfo
Tommy Atkins
80
60
40
20
0
0
0
10
20
30
40
0
10
20
30
40
Polyphenolics (mg GAE/L)
Polyphenolics (mg GAE/L)
Figure 17. Cell-growth suppressive effects of mango polyphenols on estrogen
independent MDA-MB-231 breast (A) and HT-29 colon (B) human cancer cells. Cells
were incubated for 48h with control (c) and hydrolyzed (h) phenolic extracts. Values are
mean ± SE, n=3.
The 48h incubation-effect of mango polyphenols (control) from different mango
varieties on the grown suppression of breast (IC50 = 4.7 ± 5.6 μg GAE/mL) and colon
(IC50 = 6.3 ± 4.5 μg GAE/mL) cancer cells by 50% ranged from 1 to 15 μg of gallic
94
acid equiv/mL extract (Table 10), which was equivalent to 3-132 mg of mango pulp/mL
culture medium (Table 11). Similar values were found to hydrolyzed extracts-treated
cells (Figure 17). The hydrolysis of Fr, To and Ha mango polyphenols did not statically
affect the cell proliferation of HT-29 colon and MDA-MB-231 breast cancer cells
(Table 10).
Table 10. Effect of the treatment with control and hydrolyzed mango phenolic extracts
on the growth suppression of MDA-MB-231 breast and HT-29 colon human cancer cell
lines, expressed in terms of IC50 values (mg GAE/L).
IC50 (mg GAE/L)
MDA-MB-231 (breast cancer)
HT-29 (colon cancer)
Control
Hydrolyzed
extract
Control
Hydrolyzed
extract
Tommy
Atkins
2.3 ± 0.4 b
3.5 ± 0.4 c
8.5 ± 0.7 a,b
11.5 ± 2.1 a
Ataulfo
1.1 ± 0.1 b
1.2 ± 0.5 c
7.8 ± 1.8 a,b,c*
14.3 ± 3.1 a*
Francis
2.8 ± 0.4 b
4.1 ± 1.3 b,c
12.0 ± 2.0 a
10.3 ± 0.4 a
Haden
14.6 ± 4.9 a
13.3 ± 2.5 a
0.9 ± 0.1 c
0.9 ± 0.1 b
Variety
Kent
2.5 ± 1.4 b*
8.5 ± 0.4 b*
2.5 ± 2.1 b,c
1.3 ± 0.6 b
Data are means of three or more independent determinations ± SD. Means with different letters in the
column are statistically different at p < 0.05 (Tukey-Kramer HSD test).
* Indicates statistical difference (p< 0.05) between the cancer cell growth inhibition activities of control
and hydrolyzed phenolic extract, for the respective variety and cell line (line) (paired t-student).
Table 11. Cell-growth suppressive effects of mango polyphenols (control) on MDAMB-231 breast and HT-29 colon human cancer cell lines, expressed in terms of IC50
values (mg mango pulp/mL culture medium).
Variety
Tommy Atkins
Ataulfo
Francis
Haden
Kent
IC50
(mg mango pulp/mL culture medium)
MDA-MB-231 (breast cancer)
HT-29 (colon cancer)
24 ± 4.0
84 ± 6.9
3 ± 0.3
21.6 ± 4.9
21.9 ± 3.1
93.2 ± 15.5
132 ± 44.3
8.1 ± 0.9
20.7 ± 11.6
20.7 ± 17.4
Equivalent results for the cell-growth suppressive effects of mango polyphenols
hydrolyzed or not on colon cancer cells suggest a possible contribution of βglycosidases in polyphenols enzymatic cleavage followed liberation of free active
cytotoxic aglycones that may act selectively on cancer cells, as demonstrated by Arafa
95
(2009) (52), who showed that the sensitivity degree in HT-29 and Caco-2 colon cancer
cells towards glycoconjugates over 24-96h incubation was positively correlated with βglucosidase activity assessed in these cell lines. At the same time, anticancer activity of
mango polyphenols may be also attributed to the intact polyphenols, since a reduction in
TPC of ataulfo after hydrolysis significantly decreased in 1.8-fold the effectiveness of
polyphenols in suppressing HT-29 cell growth by 50% as compared to control extracts
(Table 10), whereas Kent hydrolyzed extracts was 3.4-fold less effective in inhibiting
cell proliferation of MDA-MB-231 cells. This difference is likely associated to the
decrease in hydrolyzable tannins as a result of protein-tannin interaction, which may be
negatively
affected
the
mango
biological
activities.
Considering
that
the
chemopreventive properties of tannins have been related to their antioxidant, apoptosisinducing, and enzyme inhibitory activities (94-97), these results together suggest that
biological properties of mango polyphenols are ascribed to their bioactive metabolites
derived from enzymatic hydrolysis as well as to the parental polyphenols.
Even though some studies evaluating the chemopreventive activity of mango
pulp extracts and individual compounds have been performed (10, 19, 98), studies
regarding the potential of mango pulp phenolic fraction on colon and breast cancer are
still limited. Initially, Percival et al. (2006) (16) demonstrated that the whole mango
juice (0.01-0.1%) was effective in inhibiting chemically induced neoplastic
transformation of mammalian cell lines, BALB/3T3 cells, and also in reducing the
number of foci by 50-70% in a dose-dependent manner. Later, Prasad and others (2008)
confirmed the efficacy of aqueous mango pulp extract in inducing apoptosis in mouse
prostate (19) and in combating oxidative stress-induced cellular injury of prostate in
mice fed daily with 1mL extract (
0.2g fruit) for 15 consecutive days, by enhancing
the level of antioxidant enzymes (20). Recently, Garcia-Solis et al. (2009)
(17)
demonstrated the efficiency of mango aqueous extracts against the viability of the breast
cancer MCF-7 cell line. However, these studies failed to assess the anticancer activity of
all polyphenols present in mango pulp because aqueous extraction is not efficient in
recovering higher molecular weight tannins (99). Controversely, García-Solís, Yahia
and Aceves (2008) (18) showed that short- and long-term whole mango consumption at
physiological levels did not prevent mammary carcinogenesis in N-methyl-Nnitrosourea-treated rats, neither at the initiation nor at the promotion/progression steps
96
of carcinogenesis. In this study, the whole mango juice from the variety Ataulfo (0.020.06 g of mango/mL of water), containing carotenoids (2.3 ± 0.3 μg total
carotenoids/mL) and polyphenols (51.0 ± 3.2 μg GAE/mL) was administrated in rats at
4.5, 8.5 and 12.7 g of mango/Kg b. wt., concentrations comparable to human intake of
two (293 g of mango pulp per 70 Kg of b. wt.), four, and six mangoes per day,
respectively. Although these studies have shown the contribution of mango pulp
consumption for health benefits, biological activities of the mango phenolic fraction was
not studied individually since other bioactive compounds like L-ascorbic acid and
carotenoids were found in aqueous extracts or mango juice, respectively (17-18). These
studies underscore the great chemopreventive potential of mango phytochemicals,
especially polyphenols.
Many in vitro experiments with individual polyphenols found in mangoes have
shown antitumor-related activities, mainly in colon and breast cancer, such as induction
of apoptosis, inhibition of tumor growth and angiogenesis, and several have been
confirmed in vivo (10-15). Mangiferin has been considered the main bioactive
compound of Vimang, a industrial scale and commercially mango stem bark extract that
have been used at least 10 years in Cuba medicine with effectiveness against several
diseases, like cancer, with health benefits associated to its anti-inflammatory,
immunomodulatory and antioxidant activities (2-7). Gallic acid (GA) has been shown
significant cell growth inhibition in human cancer cell lines including esophageal,
gastric, cervix, breast MCF-7, and colon human cancer cells HT-29, Colo201 and Colon
26 (mouse colon cancer), where the GA concentration necessary to inhibit breast and
colon cancer cell growth by 50% were 0.15, 0.24, 0.18 and 0.26 mg/mL, respectively
(98). Furthermore, several studies have been reported the anticancer-related activities of
gallotannins, with potential in colon carcinogenesis (12-14, 100). 1,2,3,4,6-penta-Ogalloyl-β-D-glucose (PGG), which is the precursor of gallotannins and is present in
mango pulp, showed to induce cell cycle arrest, cell proliferation and apoptosis in a cell
type-dependent manner on human MCF breast and jurkat T cancer cells (12-13).
Additionally, treatment with gallotannins (20 μg/ml) for three days inhibited the growth
of the colon cancer cells T-84 by 50% (100). In later study, the potential molecular
mechanisms of action of gallotannins on colon cancer were evaluated in vitro. AlAyyoubi and Gali-Muhtasib (2007) (101) shown that gallotannins differentially
97
inhibited the growth of the isogenic human colon cancer cell lines HCT-116(p53 +/+)
(IC50 value
45 μg/mL) and HCT-116 (p53 -/-) (IC50 value
30 μg/mL) versus
normal human intestinal epithelial cells FHs 74Int (IC50 value > 60 μg/mL).
Indeed, previous studies have been shown that the major ability of tannins may
have a positive impact on the absorption and metabolism of polyphenols and other
constituents in human body, or may contribute to human body health benefits and
cancer prevention in vivo by complexing enzymes involved in carcinogenesis process.
On human physiology, beneficial effects of tannins may be explained by their ability to
bind proteins. These interactions may result in enzyme activity inactivation by
precipitation or by formation of soluble but inactive enzyme-inhibitor complexes and/or
by complexation with the substrate which lead to a reduction in the enzyme reaction
(102). However, mechanisms related to the effects of tannins on human food digestion
are not fully understood. Recent studies have been shown that some physiological
effects in the gut may counteract tannin antinutritional effects by increasing the
production of endogenous (digestive proteases and lipases) (103) and salivary prolinrich proteins (104) as well as biliary acids (105). In rats, pancreatic biliary juice
neutralized the inhibitory effects of grape seed tannins on brush border hydrolase
activities (105). Moreover, it has been suggested that the interaction between salivary
prolin-rich proteins and tannins contribute to form complexes which may be stable
throughout the digestion process, and this would allow the absorption and retention of
nutritionally more useful food proteins (104). Additionally, as a result of tannin-protein
interaction, tannins can act as antagonists of specific hormone receptors or inhibitors of
particular enzymes involved in carcinogenesis process (106).
Considering that gallotannins show a great contribution to the mango pulp
phenolic content, these evidences collectively suggest a potential role of mango
polyphenols as chemopreventive agents against colon and breast cancer.
98
Cell-growth supressive activity of low and high molecular weight
polyphenols-rich fraction
Hydrolyzed phenolic extract was fractionated onto C18 in high (HMW fraction)
and low molecular weight polyphenols (LMW fraction). The former represented the
major mango polyphenols derived from mango pulp consumption in their intact form,
whereas the latter correspond to low molecular weight polyphenols derived from
hydrolysis with beta-glucosidase, which are potentially absorbed through gut barrier.
HMW fraction (F1) consisted of mangiferin (422.35) and hydrolyzed tannins
with molecular weight ranged from 788 to 1851 Da, whereas LMW fraction (F2)
comprise mainly phenolic acids with molecular weight ranging from 138.12 (phydroxybenzoic acid) to 194.18 Da (ferulic acid), as characterized by HPLC-DAD
(Figure 18) and HPLC-ESI-MSn analysis (Apendice).
0.06
2.00
(A1)
8
3
1.50
0.02
3
AU
0.00
1.00
(A)
3
AU
0.04
10.00
20.00
30.00
40.00
50.00
Minutes
60.00
70.00
80.00
3
0.50
3 3 33
3 86
3
33
0.00
10.00
0.60
20.00
30.00
40.00
50.00
Minutes
60.00
70.00
80.00
(B)
60.00
70.00
80.00
78
0.40
AU
1
2
0.20
3
3
45
6
7
0.00
10.00
20.00
30.00
40.00
50.00
Minutes
Figure 18. Chromatograms at 280 nm from HMW (A) and LMW (B) fractions (F2)
from Ataulfo hydrolyzed extract. Peak assignments: (1) gallic acid; (2) p-OH-benzoic
acid; (3) gallotannins; (4) vanillic acid; (5) caffeic acid; (6) p-coumaric acid; (7) ferulic
acid. Representative chromatograms at 340 nm of HMW fraction (A1): (8) mangiferin.
99
Although HMW fraction (4308.5 mg GAE/L) presented a TPC 15-fold greater
than LMW fraction (283.4 mg GAE/L), its AA was
3-fold higher than AA from
LMW fraction (Table 12), when fractions were analyzed at the same dilution. The
higher amount of hydrolyzable tannins in HMW fraction may explain this difference.
Indeed, tannins (condensed and hydrolyzable tannins) have been shown the strongest in
vitro antioxidant activity as compared to other polyphenol structures (e.g., flavanols,
flavonols,
chalcones,
flavones,
flavanones,
isoflavones,
tannins,
stilbenes,
curcuminoids, phenolic acids, coumarins, lignans, and quinones) (31, 107). Because
tannins has relatively many hydroxyl groups and are highly polymerized, these
compounds have around 8-30 times better peroxyl radical quenching capacity than
simple phenols and Trolox (31, 99, 107), demonstrating significant scavenging action at
lower concentrations.
Table 12. Total phenolic content (TPC) and antioxidant activity of the hydrolyzed
phenolic extract and its fractions.
Fractions
Hydrolyzed phenolic extract
Antioxidant capacity
(μmol trolox eq./L)
25002.7 ± 197.6
21157.4 ± 590.7
HMW fraction
7880.4 ± 255.8
LMW fraction
Data are means of three or more independent determinations ± SD.
TPC
(mg GAE/L)
4592.0 ± 22.8
4308.5 ± 25.4
283.4 ± 14.1
Although high molecular weight polyphenols showed higher contribution to the
in vitro antioxidant capacity than the hydrolyzed phenolic extract (Table 12), the
antioxidant potential in vivo may be distant from in vitro values. Several studies have
shown that the antioxidant capacity in vivo may be related to low molecular weight
metabolites derived from microbiota transformation or from antioxidant enzymatic
system regulation and metabolism, which have antioxidant activities different from the
parental compounds. Administration of grape seed proanthocyanidin-rich extract (250
mg/kg b.w.) in rats by intragastric intubation resulted in an increase of potential
antioxidant of rat plasma, represented by the proanthocyanidins metabolites gallic acid,
(+)-catechin, and (-)-epicatechin (108). Moreover, the supplementation of a meal with
grape seed proanthocyanidins minimized the postprandial oxidative stress by decreasing
the oxidants and increasing the antioxidant levels in plasma, which enhanced the
resistance to oxidative modification of LDL (109). These results suggest that the intake
100
of tannins may increase the body resistance against oxidative stress and may contribute
to physiological functions through their in vivo antioxidative ability.
Cells were incubated with the same dilutions of polyphenols-rich fractions and
their antiproliferative activities were evaluated on MDA-MB-231 breast and HT-29
Net Growth of MDA-MB-231breast
cancer cells (% of control)
120
(A)
Net Growth of HT-29 colon cancer cells
(% of control)
colon cancer cells (Figure 19).
LMW
HMW
100
80
60
40
20
120
(B)
LMW
HMW
100
80
60
40
20
0
0
0
5
10
15
20
0
5
10
15
20
Ataulfo polyphenols (mg GAE/L)
Ataulfo polyphenols (mg GAE/L)
Figure 19. Cell-growth suppressive effects of mango polyphenols on estrogen
independent MDA-MB-231 breast (A) and human HT-29 colon (B) cancer cells. Cells
were incubated for 48h with HMW and LMW fractions from Ataulfo hydrolyzed
extract. Error bars represent the standard error of the mean (n=3).
Considering equal dilutions, HMW fraction (F1) showed better effectiveness in
inhibiting MDA-MB-231 breast and HT-29 colon cancer cell growth than LMW
fraction (F2) (Figure 20). F1 and F2 fractions equally diluted at 1: 450 (v/v) inhibited
HT-29 colon cell growth by 88.5 and 24.7 %, whereas MDA-MB-231 breast cancer cell
proliferation was inhibited by 86.9 and 1.2 %, respectively (Figure 20). In terms of
available amount of these polyphenols from the diet, these results suggest a higher
contribution of high molecular weight to health benefits derived from mango pulp
consumption, since free forms of phenolic acids are found in much lower concentrations
than HMW compounds and will likely be available in vivo after deglycosylation
process.
101
100
Dilutions
1: 900
1: 450
1: 225
(A)
80
60
40
20
0
Growth supression of HT-29 colon cancer cells
( % of control)
Growth supression of MDA-MB-231 breast cancer cells
( % of control)
120
120
100
Dilutions
1: 900
1: 450
1: 225
80
60
40
20
0
LMW
LMW
HMW
(B)
HMW
Ataulfo polyphenols (mg GAE/L)
Ataulfo polyphenols (mg GAE/L)
Figure 20. Effect of the treatment with equal dilutions of the HMW (F1) and LMW
(F2) fractions from Ataulfo hydrolyzed extract on the growth suppression of MDA-MB231 breast and HT-29 colon human cancer cell lines, expressed in percentage of control.
Error bars represent the standard error of the mean (n=3).
The great difference in cell growth suppressive effects of F1 and F2 fractions is
associated to their huge difference in TPC (15-fold) and, likely in tannin concentration,
which contribute to the higher antioxidant capacity of F1 as compared to F2 (Table 19).
On the other hand, when compared in TPC normalized to gallic acid equivalents (GAE),
both fractions were equally efficient in inhibiting the cell proliferation of MDA-MB231 breast and HT-29 colon cancer cells by 50% (Table 12). The proliferation of HT-29
colon cancer cells was inhibited in a dose-dependent manner by F1 (IC50 value = 3.0 μg
phenols /mL extract) and F2 fraction (IC50 value = 1.3 μg phenols /mL extract). Similar
efficacy was demonstrated in MDA-MB-231 cells treated with both fractions (Table 9).
Table 12. Effect of the treatment with HMW (F1) and LMW (F2) fractions on the
growth suppression of MDA-MB-231 breast and HT-29 colon human cancer cell lines,
expressed in terms of IC50 values (mg GAE/L).
IC50 (mg GAE/L)
F2 (LMW)
F1 (HMW)
Cell line
a
2.5 ± 0.0 a
4.2
±
0.1
MDA-MB-231
3.0 ± 0.0 a
1.3 ± 0.0 a
HT-29
Data are means of three independent determinations ± SD. Means with different letters in the line are
statistically different at p < 0.05 (Tukey-Kramer HSD test).
102
Therefore, these results together suggest both contribution of intact polyphenols
and their metabolites as chemopreventive compounds derived to the mango pulp
consumption. Intestinal bacteria can transform dietary polyphenols into a wide range of
low molecular weight compounds that are potentially more biologically active than the
parent compounds (23, 25-27) (110). The bioactive ferulic acid, for example, may be
released from its ethyl ester for the action of the bacteria found in human feces,
including Bifidobacterium lactis, Lactobacillus gassseri and Escherichia coli (111).
Urolithins, the metabolites derived from ellagitannins degradation by gut microflora,
have been reported to exert biological activity (112-114), although the microbiota
involved in their production was not identified so far. Additionally, proanthocyanidins
from cranberries showed to be effective in reducing the risk of recurrent urinary tract
infections by reducing adhesion of bladder cells (115-116). Russel et al. (2207) (117)
demonstrated that cytokine-induced stimulation of the inflammatory pathways in colon
cells was four-fold up-regulated in the presence of the free phenolic acid fraction from
blueberries, suggesting that health benefits of blueberry phenolics as anti-inflammatory
agents in the colon are likely resulted of microbial metabolism. Moreover, because of
the limited absorption of high molecular weight polyphenols through intestinal gut
barrier, substantial levels of unabsorbed dietary polyphenols in large intestine. These
polyphenols may modulate the colonic microbiota by increasing e/or reducing the
number of specific microbial strains and thus enhancing colon health. Tea polyphenols
including epicathechin, catechin, gallic acid, caffeic acid and 3-O-methylgallic acid
showed to repress the growth of Clostridium perfringens, Clostridium difficile and
Bacteroides spp., whereas the probiotics Bifidobacterium spp. and Lactobacillus spp.
were less affected (110). Tannin-rich berries have been shown to exhibit antimicrobial
properties against pathogenic bacteria, which may contribute to improve colon health
(118). A lactobacilli-supplemented diet in rats treated with the colon carcinogen 1,2dimethylhydrazine hydrochloride (DMH) induced a decrease in the extent of DMHinduced DNA damage, especially in rat colon cells. The chemopreventive effects
associated to the supplementation with Lactobacilli casei were accompanied by changes
in the activities of several xenobiotic metabolizing enzymes, where the concentrations
of reduced glutathione and the activities of glutathione S-transferase, glutathione
peroxidase and superoxide dismutase were down-regulated as a response to an
103
improved antioxidant status(119). According to these evidences, both parental
polyphenols as well as their metabolites derived from gut microflora transformation
may reduce oxidative stress, inflammation, pathogenic bacteria, and alter the balance
between the bio-activation and detoxification metabolic pathways by inducing the
production of enzymes. These beneficial effects derived from polyphenols consumption
might conjunctly reduce the risk and/or protect against colon carcinogenesis by enhance
colon health (120).
Indeed, even though the occurrence of hydrolyzable tannins (mango,
pomegranate, berries) is much more limited that that of condensed tannins (tea, wine,
muscadine grapes, berries) (121), several investigations have suggested that dietary
administration of hydrolyzable tannins-rich foods inhibited events associated with both
the initiation and promotion/progression of cancer in vivo (122-123). Blackberry,
raspberry, and strawberry are known to contain predominantly hydrolyzable tannins
(ellagitannins and gallotannins), whereas blueberries and cranberries are known to
contain predominantly condensed tannins (proanthocyanidins) (124). Seeram et al.
(2006) (125) showed that phenolic extracts from blackberry (IC50 value = 64.60 μg/mL),
blueberry (89.96 μg/mL) and black raspberry (89.11 μg/mL) shown to inhibit the
growth of HT-29 colon cancer cells by 50% more efficiently than strawberry (114.20
μg/mL), cranberry (121.30 μg/mL), and red raspberry (187.60 μg/mL) extracts. In later
study, polyphenol-rich berry extracts were screened for their antiproliferative
effectiveness using Caco-2 human colon cancer cells. The viability of Caco-2 colon
cancer cells was inhibited in a dose-dependent manner by strawberry (IC50 value = 25.5
μg phenols /mL extract), arctic bramble (26.4 μg /mL), cloudberry (31.6 μg /mL) and
lingonberry (28.7 μg /mL) phenolic extracts. Interestingly, the antiproliferative activity
of procyanidins -rich fraction from lingonberry extracts was more effective than the
original extract and anthocyanin-rich fraction (126). In similar study, Ross, McDougall
and Stewart (2007) (127) shown that the antiproliferative activity of raspberry phenolic
extracts is predominantly associated to ellagitannins instead of anthocyanins, because
ellagitannins-rich fraction (IC50 = 13.0 μg GAE/ml) was more effective in inhibiting
human cervical cancer (HeLa) cell growth by 50% than whole raspberry phenolic
extract (17.5 μg GAE/ml) and anthocyanin-enriched fraction (67.0 μg GAE/ml).
Tannin-rich fractions (85.1- 93.2% of the TPC) from four varieties of muscadine grapes,
104
which contained predominantly hydrolyzable tannins (128), showed to inhibit the cell
proliferation of HT-29 (
300 μg/mL) and Caco-2 (300-500 μg/mL) colon cancer cell
lines, showing a 2-4 greater inhibitory activity as compared to phenolic acid fraction
(129).
In addition, the degree of galloylation may play a role in the cytotoxic effects in
colon cancer cells (130-131), which may indicate that some of the anti-cancer effects
may be influenced by the high degree of galloylation of mango-polyphenols. Tannins
which exhibited potent inhibitory activity on the tumor cell motility have common
characteristics such as galloyl groups substituting all the hydroxyl groups of β-Dglucose and some of them cross-linked to form hexahydroxydiphenoyl. Ellagitannins
including the most potent 1,2,4-tri-O-galloyl-3,6-hexahydroxydiphenoyl-β-D-glucose
(punicafolin), showed to be more potent than gallotannins (mono, di and penta-Ogalloyl- β-D-glucose) (132). In similar study, Coriariin A, which has four galloyl
groups, showed higher antitumor than agrimoniin, that has two hexahydroxydiphenol
groups (32). This results are in agreement with Lizarra et al. (2008) (131) findings. The
more galloylated Witch hazel fractions showed better effectiveness at inhibiting
proliferation of HT-29 and HCT-116 human colon cancer cells, at arresting the cell
cycle at the S phase as well as at inducing apoptosis and necrosis. Interestingly, the
apoptosis and cell cycle arrest effects were proportional to their galloylation level.
Moreover, witch hazel fractions with a high degree of galloylation were also the most
effective as scavengers of both hydroxyl and superoxide radicals and in protecting DNA
damage triggered by the hydroxyl radical system. These evidences suggest
chemopreventive effects of mango gallotannins, which present high level of
galloyllation degree, varying from one to eleven galloyl moieties attached in a glucose
core.
Conclusion
The absorption of mango polyphenols was predominantly characterized by low
molecular weight compounds, mainly phenolic acids in their free forms like gallic,
caffeic, ferulic, p-OH- coumaric and vanilic acids, with no evidence of absorption of
high molecular weight polyphenols like mangiferin (422 Da) and hydrolyzable tannins
105
with molecular weight ranging from 788 Da (tetra-O-galloyl-glucose) to 1852 Da
(undeca-O-galloyl-glucose). Despite the enzymatic hydrolysis had enhanced the mango
polyphenols absorption through Caco-2 intestinal monolayers, in general, mango
polyphenols hydrolysis with β-glucosidase activity did not increased total phenolic
content, antioxidant activity and growth suppression of MDA-MB-231 breast and HT29 colon cancer cells. Additionally, both high and low molecular weight polyphenols
fractions equally inhibited cell proliferation of colon and breast cancer cells at the same
extent (0-20 μg of gallic acid equiv/mL). These results suggest that the most of mango
polyphenols are not absorbed intact in vivo through the small intestine, but may be
hydrolyzed by intestinal enzymes into low molecular weight aromatic acids, which are
later absorbed, or likely reach the large intestine, modulating the gut microflora and
protecting colon carcinogenesis. It suggests that the anti-cancer effects of mango
polyphenols may not require their absorption in vivo through the gut barrier. Additional
studies regarding the chemopreventive mechanisms are necessary to better elucidate the
anti-cancer properties of mango polyphenols.
106
REFERENCES
1.
FAOSTAT, FAO Statistical Database – Agriculture.
http://apps.fao.org. Accessed: September, 2009.
2.
Garrido, G.; Gonzalez, D.; Delporte, C.; Backhouse, N.; Quintero, G.; Nunez-
Selles, A. J.; Morales, M. A., Analgesic and anti-inflammatory effects of Mangifera
indica L. extract (Vimang). Phytother Res 2001, 15, (1), 18-21.
3.
Garrido, G.; Gonzalez, D.; Lemus, Y.; Garcia, D.; Lodeiro, L.; Quintero, G.;
Delporte, C.; Nunez-Selles, A. J.; Delgado, R., In vivo and in vitro anti-inflammatory
activity of Mangifera indica L. extract (VIMANG). Pharmacol Res 2004, 50, (2), 143-9.
4.
Garrido, G.; Delgado, R.; Lemus, Y.; Rodriguez, J.; Garcia, D.; Nunez-Selles, A.
J., Protection against septic shock and suppression of tumor necrosis factor alpha and
nitric oxide production on macrophages and microglia by a standard aqueous extract of
Mangifera indica L. (VIMANG). Role of mangiferin isolated from the extract.
Pharmacol Res 2004, 50, (2), 165-72.
5.
Martinez, G.; Delgado, R.; Perez, G.; Garrido, G.; Selles, A. J. N.; Leon, O. S.,
Evaluation of the in vitro antioxidant activity of Mangifera indica L. extract (Vimang).
Phytother Res 2000, 14, (6), 424-427.
6.
Nunez-Selles, A. J., Antioxidant therapy: Myth or reality? J Brazil Chem Soc
2005, 16, (4), 699-710.
7.
Nunez-Selles, A. J.; Delgado-Hernandez, R.; Garrido-Garrido, G.; Garcia-
Rivera, D.; Guevara-Garcia, M.; Pardo-Andreu, G. L., The paradox of natural products
as pharmaceuticals - Experimental evidences of a mango stem bark extract. Pharmacol
Res 2007, 55, (5), 351-358.
8.
Schieber, A.; Ullrich, W.; Carle, R., Characterization of polyphenols in mango
puree concentrate by HPLC with diode array and mass spectrometric detection
Innovative Food Science and Emerging Technologies 2000, 1, (2), 161-166.
9.
Berardini, N.; Carle, R.; Schieber, A., Characterization of gallotannins and
benzophenone derivatives from mango (Mangifera indica L. cv. 'Tommy Atkins') peels,
pulp and kernels by high-performance liquid chromatography electrospray ionization
mass spectrometry. Rapid Commun Mass Sp 2004, 18, (19), 2208-2216.
107
10.
Huh, J. E.; Lee, E. O.; Kim, M. S.; Kang, K. S.; Kim, C. H.; Cha, B. C.; Surh, Y.
J.; Kim, S. H., Penta-O-galloyl-beta-D-glucose suppresses tumor growth via inhibition
of angiogenesis and stimulation of apoptosis: roles of cyclooxygenase-2 and mitogenactivated protein kinase pathways. Carcinogenesis 2005, 26, (8), 1436-1445.
11.
Zhang, J. H.; Li, L.; Kim, S. H.; Hagerman, A. E.; Lu, J. X., Anti-Cancer, Anti-
Diabetic and Other Pharmacologic and Biological Activities of Penta-Galloyl-Glucose.
Pharm Res-Dord 2009, 26, (9), 2066-2080.
12.
Chen, W. J.; Chang, C. Y.; Lin, J. K., Induction of G1 phase arrest in MCF
human breast cancer cells by pentagalloylglucose through the down-regulation of CDK4
and CDK2 activities and up-regulation of the CDK inhibitors p27(Kip) and p21(Cip).
Biochemical Pharmacology 2003, 65, (11), 1777-1785.
13.
Chen, W. J.; Lin, J. K., Induction of G1 arrest and apoptosis in human jurkat T
cells by pentagalloylglucose through inhibiting proteasome activity and elevating
p27Kip1, p21Cip1/WAF1, and Bax proteins. J Biol Chem 2004, 279, (14), 13496-505.
14.
Oh, G. S.; Pae, H. O.; Oh, H.; Hong, S. G.; Kim, I. K.; Chai, K. Y.; Yun, Y. G.;
Kwon, T. O.; Chung, H. T., In vitro anti-proliferative effect of 1,2,3,4,6-penta-Ogalloyl-beta-D-glucose on human hepatocellular carcinoma cell line, SK-HEP-1 cells.
Cancer Lett 2001, 174, (1), 17-24.
15.
Liu, Z. J.; Schwimer, J.; Liu, D.; Lewis, J.; Greenway, F. L.; York, D. A.;
Woltering, E. A., Gallic acid is partially responsible for the antiangiogenic activities of
Rubus leaf extract. Phytother Res 2006, 20, (9), 806-813.
16.
Percival, S. S.; Talcott, S. T.; Chin, S. T.; Mallak, A. C.; Lounds-Singleton, A.;
Pettit-Moore, J., Neoplastic transformation of BALB/3T3 cells and cell cycle of HL-60
cells are inhibited by mango (Mangifera indica L.) juice and mango juice extracts.
Journal of Nutrition 2006, 136, (5), 1300-1304.
17.
Garcia-Solis, P.; Yahia, E. M.; Morales-Tlalpan, V.; Diaz-Munoz, M., Screening
of antiproliferative effect of aqueous extracts of plant foods consumed in Mexico on the
breast cancer cell line MCF-7. Int J Food Sci Nutr 2009, 1-15.
18.
Garcia-Solis, P.; Yahia, E. M.; Aceves, C., Study of the effect of 'Ataulfo'
mango (Mangifera indica L.) intake on mammary carcinogenesis and antioxidant
capacity in plasma of N-methyl-N-nitrosourea (MNU)-treated rats. Food Chem 2008,
111, (2), 309-315.
108
19.
Prasad, S.; Kalra, N.; Shukla, Y., Induction of apoptosis by lupeol and mango
extract in mouse prostate and LNCaP cells. Nutr Cancer 2008, 60, (1), 120-30.
20.
Prasad, S.; Kalra, N.; Singh, M.; Shukla, Y., Protective effects of lupeol and
mango extract against androgen induced oxidative stress in Swiss albino mice. Asian J
Androl 2008, 10, (2), 313-8.
21.
Scalbert, A.; Morand, C.; Manach, C.; Remesy, C., Absorption and metabolism
of polyphenols in the gut and impact on health. Biomed Pharmacother 2002, 56, (6),
276-282.
22.
Berrin, J. G.; McLauchlan, W. R.; Needs, P.; Williamson, G.; Puigserver, A.;
Kroon, P. A.; Juge, N., Functional expression of human liver cytosolic beta-glucosidase
in Pichia pastoris. Insights into its role in the metabolism of dietary glucosides. Eur J
Biochem 2002, 269, (1), 249-58.
23.
Selma, M. V.; Espin, J. C.; Tomas-Barberan, F. A., Interaction between
Phenolics and Gut Microbiota: Role in Human Health. J Agr Food Chem 2009, 57, (15),
6485-6501.
24.
Cerda, B.; Llorach, R.; Ceron, J. J.; Espin, J. C.; Tomas-Barberan, F. A.,
Evaluation of the bioavailability and metabolism in the rat of punicalagin, an
antioxidant polyphenol from pomegranate juice. European Journal of Nutrition 2003,
42, (1), 18-28.
25.
Li, M. S.; Yao, K.; He, Q.; Jia, D. Y., Biodegradation of gallotannins and
ellagitannins. J Basic Microb 2006, 46, (1), 68-84.
26.
Deprez, S.; Mila, I.; Huneau, J. F.; Tome, D.; Scalbert, A., Transport of
proanthocyanidin dimer, trimer, and polymer across monolayers of human intestinal
epithelial Caco-2 cells. Antioxid Redox Sign 2001, 3, (6), 957-967.
27.
Scalbert, A.; Williamson, G., Dietary intake and bioavailability of polyphenols.
Journal of Nutrition 2000, 130, (8), 2073s-2085s.
28.
Swain, T.; Hillis, W., The phenolic constituents of Prinus domestica. I. The
Quantitative analysis of phenolic constituents. . Journal of the Science and Food Agric
1959, 10, 63-68.
29.
Lafay, S.; Gil-Izquierdo, A., Bioavailability of phenolic acids Phytochemistry
Reviews 2008, 7, (2), 301-311.
109
30.
Nemeth, K.; Plumb, G. W.; Berrin, J. G.; Juge, N.; Jacob, R.; Naim, H. Y.;
Williamson, G.; Swallow, D. M.; Kroon, P. A., Deglycosylation by small intestinal
epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of
dietary flavonoid glycosides in humans. Eur J Nutr 2003, 42, (1), 29-42.
31.
Cai, Y. Z.; Mei, S.; Jie, X.; Luo, Q.; Corke, H., Structure-radical scavenging
activity relationships of phenolic compounds from traditional Chinese medicinal plants.
Life Sci. 2006, 78, (25), 2872-88.
32.
Miyamoto, K.; Kishi, N.; Koshiura, R.; Yoshida, T.; Hatano, T.; Okuda, T.,
Relationship between the Structures and the Antitumor Activities of Tannins. Chem
Pharm Bull 1987, 35(2) (2), 814-822.
33.
Talcott, S. T.; Lee, J. H., Ellagic acid and flavonoid antioxidant content of
muscadine wine and juice. J Agric Food Chem 2002, 50, (11), 3186-92.
34.
Hidalgo, I. J.; Raub, T. J.; Borchardt, R. T., Characterization of the human colon
carcinoma cell line (Caco-2) as a model system for intestinal epithelial permeability.
Gastroenterology 1989, 96, (3), 736-49.
35.
Ribeiro, S. M. R.; Queiroz, J. H.; Queiroz, M. E. L. R. d.; Campos, F. M.;
Sant’Ana, H. M. P., Antioxidant in mango (Mangifera indica L.) pulp Plant Food Hum
Nutr 2007, 62, (1), 13-17.
36.
Kim, Y.; Brecht, J. K.; Talcott, S. T., Antioxidant phytochemical and fruit
quality changes in mango (Mangifera indica L.) following hot water immersion and
controlled atmosphere storage. Food Chem 2007, 105, (4), 1327-1334.
37.
Ribeiro, S. M. R.; Barbosa, L. C. A.; Queiroz, J. H.; Knodler, M.; Schieber, A.,
Phenolic compounds and antioxidant capacity of Brazilian mango (Mangifera indica L.)
varieties. Food Chem 2008, 110, (3), 620-626.
38.
Wu, X. L.; Beecher, G. R.; Holden, J. M.; Haytowitz, D. B.; Gebhardt, S. E.;
Prior, R. L., Lipophilic and hydrophilic antioxidant capacities of common foods in the
United States. J Agr Food Chem 2004, 52, (12), 4026-4037.
39.
Kawamoto, H.; Nakatsubo, F.; Murakami, K., Stoichiometric studies of tannin-
protein co-precipitation. Phytochemistry 1996, 41, (5), 1427-1431.
40.
Dewick, P. M., The Biosynthesis of Shikimate Metabolites. Nat Prod Rep 1993,
10, (3), 233-263.
110
41.
Niemetz, R.; Gross, G. G., Gallotannin biosynthesis: beta-glucogallin:
hexagalloyl 3-O-galloyltransferase from Rhus typhina leaves. Phytochemistry 2001, 58,
(5), 657-661.
42.
He, Q.; Shi, B.; Yao, K., Interactions of gallotannins with proteins, amino acids,
phospholipids and sugars
Food Chemistry 2006, 95, (2), 250-254.
43.
Hagerman, A. E.; Butler, L. G., The Specificity of Proanthocyanidin-Protein
Interactions. Journal of Biological Chemistry 1981, 256, (9), 4494-4497.
44.
Butler, L. G.; Riedl, D. J.; Lebryk, D. G.; Blytt, H. J., Interaction of Proteins
with Sorghum Tannin - Mechanism, Specificity and Significance. J Am Oil Chem Soc
1984, 61, (5), 916-920.
45.
Sarni-Manchado, P.; Canals-Bosch, J. M.; Mazerolles, G.; Cheynier, V.,
Influence of the glycosylation of human salivary proline-rich proteins on their
interactions with condensed tannins. J Agric Food Chem 2008, 56, (20), 9563-9.
46.
Czjzek, M.; Cicek, M.; Zamboni, V.; Bevan, D. R.; Henrissat, B.; Esen, A., The
mechanism of substrate (aglycone) specificity in beta-glucosidases is revealed by crystal
structures of mutant maize beta-glucosidase-DIMBOA, -DIMBOAGlc, and -dhurrin
complexes. P Natl Acad Sci USA 2000, 97, (25), 13555-13560.
47.
Henrissat, B.; Callebaut, I.; Fabrega, S.; Lehn, P.; Mornon, J. P.; Davies, G.,
Conserved Catalytic Machinery and the Prediction of a Common Fold for Several
Families of Glycosyl Hydrolases. P Natl Acad Sci USA 1995, 92, (15), 7090-7094.
48.
Davies, G.; Henrissat, B., Structures and mechanisms of glycosyl hydrolases.
Structure 1995, 3, (9), 853-9.
49.
Osborne, N. J. T.; McNeill, D. M., Characterisation of Leucaena
condensed tannins by size and protein precipitation capacity. . J.Sci. Food Agric. 2001,
81, 1113-1119.
50.
Hagerman,
A.
E.,
Tannin
Chemistry.
<http://www.users.muohio.edu/hagermae/> 2009.
51.
Glew, R. H.; Gopalan, V.; Forsyth, G. W.; Vander Jagt, D. J., In: β-
Glucosidases: Biochemistry and Molecular Biology,
ACS, Washington, DC. 1993, 83-112.
111
52.
Arafa, H. M., Possible contribution of beta-glycosidases and caspases in the
cytotoxicity of novel glycoconjugates in colon cancer cells. Invest New Drugs 2009.
53.
Mizuma, T.; Fuseda, N.; Hayashi, M., Kinetic characterization of glycosidase
activity from disaccharide conjugate to monosaccharide conjugate in Caco-2 cells. J
Pharm Pharmacol 2005, 57, (5), 661-4.
54.
Lambert, N.; Kroon, P. A.; Faulds, C. B.; Plumb, G. W.; McLauchlan, W. R.;
Day, A. J.; Williamson, G., Purification of cytosolic beta-glucosidase from pig liver and
its reactivity towards flavonoid glycosides. Biochim Biophys Acta 1999, 1435, (1-2),
110-6.
55.
Henry, C.; Vitrac, X.; Decendit, A.; Ennamany, R.; Krisa, S.; Merillon, J. M.,
Cellular uptake and efflux of trans-piceid and its aglycone trans-resveratrol on the apical
membrane of human intestinal Caco-2 cells. J Agric Food Chem 2005, 53, (3), 798-803.
56.
Higdon, J. V.; Frei, B., Tea catechins and polyphenols: Health effects,
metabolism, and antioxidant functions. Crit Rev Food Sci 2003, 43, (1), 89-143.
57.
Rios, L. Y.; Bennett, R. N.; Lazarus, S. A.; Remesy, C.; Scalbert, A.;
Williamson, G., Cocoa procyanidins are stable during gastric transit in humans'. Am J
Clin Nutr 2002, 76, (5), 1106-1110.
58.
General, T.; Mamatha, V.; Divya, V.; Appaiah, K. A. A., Diversity of yeast with
beta-glycosidase activity in vanilla (Vanilla planifolia) plant. Curr Sci India 2009, 96,
(11), 1501-1505.
59.
Lee, M. H.; Chen, R. L. C.; Matsumoto, K., Novel enzymatic assays for the
measurement of gallotannin, amino acids, and glutamate in green tea infusions:
Analytical results. Biosci Biotech Bioch 1996, 60, (2), 362-363.
60.
Lekha, P. K.; Lonsane, B. K., Production and application of tannin acyl
hydrolase: State of the art. Adv Appl Microbiol 1997, 44, 215-260.
61.
Bhat, T. K.; Singh, B.; Sharma, O. P., Microbial degradation of tannins - A
current perspective. Biodegradation 1998, 9, (5), 343-357.
62.
Spencer, J. P. E.; Abd El Mohsen, M.; Minihane, A. M., Metabolism of dietary
phytochemicals: A review of the metabolic forms identified in humans. Curr Top
Nutraceut R 2006, 4, (3-4), 187-203.
63.
Nowak, A.; Libudzisz, Z., Carcinogenic Activity of Intestinal Microbiota. Zywn-
Nauk Technol Ja 2008, 15, (6), 25-39.
112
64.
Konishi, Y.; Kobayashi, S.; Shimizu, M., Transepithelial transport of p-coumaric
acid and gallic acid in Caco-2 cell monolayers. Biosci Biotechnol Biochem 2003, 67,
(11), 2317-24.
65.
Vaidyanathan, J. B.; Walle, T., Transport and metabolism of the tea flavonoid (-
)-epicatechin by the human intestinal cell line Caco-2. Pharm Res 2001, 18, (10), 14205.
66.
Deprez, S.; Mila, I.; Huneau, J. F.; Tome, D.; Scalbert, A., Transport of
proanthocyanidin dimer, trimer, and polymer across monolayers of human intestinal
epithelial Caco-2 cells. Antioxid Redox Signal 2001, 3, (6), 957-67.
67.
Konishi, Y.; Hagiwara, K.; Shimizu, M., Transepithelial transport of fluorescein
in caco-2 cell monolayers and use of such transport in in vitro evaluation of phenolic
acid availability. Biosci Biotech Bioch 2002, 66, (11), 2449-2457.
68.
Konishi, Y.; Shimizu, M., Transepithelial transport of ferulic acid by
monocarboxylic acid transporter in Caco-2 cell monolayers. Biosci Biotechnol Biochem
2003, 67, (4), 856-62.
69.
Konishi, Y.; Kobayashi, S., Microbial metabolites of ingested caffeic acid are
absorbed by the monocarboxylic acid transporter (MCT) in intestinal Caco-2 cell
monolayers. J Agric Food Chem 2004, 52, (21), 6418-24.
70.
Konishi, Y.; Kobayashi, S., Transepithelial transport of chlorogenic acid, caffeic
acid, and their colonic metabolites in intestinal caco-2 cell monolayers. J Agric Food
Chem 2004, 52, (9), 2518-26.
71.
Konishi, Y.; Hitomi, Y.; Yoshioka, E., Intestinal absorption of p-coumaric and
gallic acids in rats after oral administration. J Agric Food Chem 2004, 52, (9), 2527-32.
72.
Shahrzad, S.; Bitsch, I.; , Determination of gallic acid and its
metabolites in human plasma and urine by high-performance
liquid chromatography. J Chromatogr B 1998, 705, 87–95.
73.
Shahrzad, S.; Aoyagi, K.; Winter, A.; Koyama, A.; Bitsch, I., Pharmacokinetics
of gallic acid and its relative bioavailability
from tea in healthy humans. J Nutr 2001, 131, 1207-1210.
74.
Scholz, S.; Williamson, G., Interactions affecting the bioavailability of dietary
polyphenols in vivo. Int J Vitam Nutr Res 2007, 77, (3), 224-35.
113
75.
Adam, A.; Crespy, V.; Levrat-Verny, M. A.; Leenhardt, F.; Leuillet, M.;
Demigne, C.; Remesy, C., The bioavailability of ferulic acid is governed primarily by
the food matrix rather than its metabolism in intestine and liver in rats. Journal of
Nutrition 2002, 132, (7), 1962-1968.
76.
Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J., Experimental and
computational approaches to estimate solubility and permeability in drug discovery and
development settings. Adv Drug Deliver Rev 2001, 46, (1-3), 3-26.
77.
Clark, D. E., Rapid calculation of polar molecular surface area and its
application to the prediction of transport phenomena. 1. Prediction of intestinal
absorption. J Pharm Sci 1999, 88, (8), 807-814.
78.
Lafay, S.; Morand, C.; Manach, C.; Besson, C.; Scalbert, A., Absorption and
metabolism of caffeic acid and chlorogenic acid in the small intestine of rats. Br J Nutr
2006, 96, (1), 39-46.
79.
Konishi, Y.; Zhao, Z. H.; Shimizu, M., Phenolic acids are absorbed from the rat
stomach with different absorption rates. J Agr Food Chem 2006, 54, (20), 7539-7543.
80.
Zhao, Z. H.; Egashira, Y.; Sanada, H., Ferulic acid is quickly absorbed from rat
stomach as the free form and then conjugated mainly in liver. Journal of Nutrition 2004,
134, (11), 3083-3088.
81.
Bourne, L.; Paganga, G.; Baxter, D.; Hughes, P.; Rice-Evans, C., Absorption of
ferulic acid from low-alcohol beer. Free Radic Res 2000, 32, (3), 273-80.
82.
Anson, N. M.; van den Berg, R.; Havenaar, R.; Bast, A.; Haenen, G. R. M. M.,
Bioavailability of ferulic acid is determined by its bioaccessibility. J Cereal Sci 2009,
49, (2), 296-300.
83.
Bourne, L. C.; Rice-Evans, C., Bioavailability of ferulic acid. Biochem Bioph
Res Co 1998, 253, (2), 222-227.
84.
Bourne, L. C.; Rice-Evans, C. A., Detecting and measuring bioavailability of
phenolics and flavonoids in humans: Pharmacokinetics of urinary excretion of dietary
ferulic acid. Oxidants and Antioxidants, Pt A 1999, 299, 91-106.
85.
Azuma, K.; Ippoushi, K.; Nakayama, M.; Ito, H.; Higashio, H.; Terao, J.,
Absorption of chlorogenic acid and caffeic acid in rats after oral administration. J Agric
Food Chem 2000, 48, (11), 5496-500.
114
86.
Uang, Y. S.; Kang, F. L.; Hsu, K. Y., Determination of caffeic acid in rabbit
plasma by high-performance liquid chromatography. J Chromatogr B Biomed Appl
1995, 673, (1), 43-9.
87.
Yang, C. S.; Landau, J. M.; Huang, M. T.; Newmark, H. L., Inhibition of
carcinogenesis by dietary polyphenolic compounds. Annu Rev Nutr 2001, 21, 381-406.
88.
Adams, L. S.; Seeram, N. P.; Aggarwal, B. B.; Takada, Y.; Sand, D.; Heber, D.,
Pomegranate juice, total pomegranate ellagitannins, and punicalagin suppress
inflammatory cell signaling in colon cancer cells. J Agric Food Chem 2006, 54, (3),
980-5.
89.
Mertens-Talcott, S. U.; Jilma-Stohlawetz, P.; Rios, J.; Hingorani, L.; Derendorf,
H., Absorption, metabolism, and antioxidant effects of pomegranate (Punica granatum
L.) polyphenols after ingestion of a standardized extract in healthy human volunteers. J
Agr Food Chem 2006, 54, (23), 8956-8961.
90.
Larrosa, M.; Tomas-Barberan, F. A.; Espin, J. C., The dietary hydrolysable
tannin punicalagin releases ellagic acid that induces apoptosis in human colon
adenocarcinoma Caco-2 cells by using the mitochondrial pathway. J Nutr Biochem
2006, 17, (9), 611-25.
91.
Rosazza, J. P.; Huang, Z.; Dostal, L.; Volm, T.; Rousseau, B., Review:
biocatalytic transformations of ferulic acid: an abundant aromatic natural product. J Ind
Microbiol 1995, 15, (6), 457-71.
92.
Mathew,
S.;
Abraham,
T.
E.,
Bioconversions
of
ferulic
acid,
an
hydroxycinnamic acid. Crit Rev Microbiol 2006, 32, (3), 115-25.
93.
Yang, C. S.; Sang, S.; Lambert, J. D.; Lee, M. J., Bioavailability issues in
studying the health effects of plant polyphenolic compounds. Mol Nutr Food Res 2008,
52 Suppl 1, S139-51.
94.
Santos-Buelga, C.; Scalbert, A., Proanthocyanidins and tannin-like compounds -
nature, occurrence, dietary intake and effects on nutrition and health. J Sci Food Agr
2000, 80, (7), 1094-1117.
95.
Jo, J. Y.; Lee, C. Y., Cancer chemoprevention by dietary proanthocyanidins.
Food Sci Biotechnol 2007, 16, (4), 501-508.
96.
Aruoma, O. I.; Sun, B. X.; Fujii, H.; Neergheen, V. S.; Bahorun, T.; Kang, K. S.;
Sung, M. K., Low molecular proanthocyanidin dietary biofactor Oligonol: Its
115
modulation of oxidative stress, bioefficacy, neuroprotection, food application and
chemoprevention potentials. Biofactors 2006, 27, (1-4), 245-265.
97.
Jeong, W. S.; Kong, A. N. T., Biological properties of monomeric and polymeric
catechins: Green tea catechins and procyanidins. Pharm Biol 2004, 42, 84-93.
98.
Faried, A.; Kurnia, D.; Faried, L. S.; Usman, N.; Miyazaki, T.; Kato, H.;
Kuwano, H., Anticancer effects of gallic acid isolated from Indonesian herbal medicine,
Phaleria macrocarpa (Scheff.) Boerl, on human cancer cell lines. Int J Oncol 2007, 30,
(3), 605-13.
99.
Hagerman, A. E., Extraction of tannin from fresh and preserved leaves Journal
of Chemical Ecology 1988, 14, (2), 453-461.
100.
Gali-Muhtasib, H. U.; Younes, I. H.; Karchesy, J. J.; El-Sabban, M. E., Plant
tannins inhibit the induction of aberrant crypt foci and colonic tumors by 1,2dimethylhydrazine in mice. Nutrition and Cancer-an International Journal 2001, 39,
(1), 108-116.
101.
Al-Ayyoubi, S.; Gali-Muhtasib, H., Differential apoptosis by gallotannin in
human colon cancer cells with distinct p53 status. Mol Carcinog 2007, 46, (3), 176-86.
102.
Haslam, E., Plant polyphenols. Vegetable tannins revised. ed.; Cambridge
University Press: Cambridge,U.K. 1989.
103.
Tebib, K.; Besancon, P.; Rouanet, J. M., Dietary Grape Seed Tannins Affect
Lipoproteins, Lipoprotein Lipases and Tissue-Lipids in Rats Fed Hypercholesterolemic
Diets. Journal of Nutrition 1994, 124, (12), 2451-2457.
104.
Mole, S.; Butler, L. G.; Iason, G., Defense against Dietary Tannin in Herbivores
- a Survey for Proline Rich Salivary Proteins in Mammals. Biochem Syst Ecol 1990, 18,
(4), 287-293.
105.
Tebib, K.; Rouanet, J. M.; Besancon, P., Effect of Grape Seed Tannins on the
Activity of Some Rat Intestinal Enzyme-Activities. Enzyme Protein 1994, 48, (1), 5160.
106.
Polya, G. M., Biochemical targets of plant bioactive compounds: a
pharmacological 2003, 847.
107.
Yokozawa, T.; Chen, C. P.; Dong, E.; Tanaka, T.; Nonaka, G. I.; Nishioka, I.,
Study on the inhibitory effect of tannins and flavonoids against the 1,1-diphenyl-2picrylhydrazyl radical. Biochemical Pharmacology 1998, 56, (2), 213-222.
116
108.
Koga, T.; Moro, K.; Nakamori, K.; Yamakoshi, J.; Hosoyama, H.; Kataoka, S.;
Ariga, T., Increase of antioxidative potential of rat plasma by oral administration of
proanthocyanidin-rich extract from grape seeds. J Agr Food Chem 1999, 47, (5), 18921897.
109.
Natella, F.; Belelli, F.; Gentili, V.; Ursini, F.; Scaccini, C., Grape seed
proanthocyanidins prevent plasma postprandial oxidative stress in humans. J Agr Food
Chem 2002, 50, (26), 7720-7725.
110.
Lieberman, S., Are the differences between estradiol and other estrogens,
naturally occurring or synthetic, merely semantical? Journal of Clinical Endocrinology
& Metabolism 1996, 81, (2), 850-851.
111.
Couteau, D.; McCartney, A. L.; Gibson, G. R.; Williamson, G.; Faulds, C. B.,
Isolation and characterization of human colonic bacteria able to hydrolyse chlorogenic
acid. J Appl Microbiol 2001, 90, (6), 873-881.
112.
Larrosa, M.; Gonzalez-Sarrias, A.; Garcia-Conesa, M. T.; Tomas-Barberan, F.
A.; Espin, J. C., Urolithins, ellagic acid-derived metabolites produced by human colonic
microflora, exhibit estrogenic and antiestrogenic activities. J Agr Food Chem 2006, 54,
(5), 1611-1620.
113.
Seeram, N. P.; Aronson, W. J.; Zhang, Y.; Henning, S. M.; Moro, A.; Lee, R. P.;
Sartippour, N.; Harris, D. M.; Rettig, M.; Suchard, M. A.; Pantuck, A. J.; Belldegrun,
A.; Heber, D., Pomegranate ellagitannin-derived metabolites inhibit prostate cancer
growth and localize to the mouse prostate gland. J Agr Food Chem 2007, 55, (19),
7732-7737.
114.
Gonzalez-Sarrıas, A.; Espın, J. C.; Tomas-Barbera, F. A.; Garcıa-Conesa, M. T.,
Comparative transcriptional analysis reveals key cell cycle and MAPK signalling genes
involved in the S-G2/Mphase arrest of Caco-2 cells exposed to ellagic acid and its
colonic derivatives, urolithins. Mol. Nut. Food Res. 2009, 53, (686–698).
115.
Reid, G.; Hsiehl, J.; Potter, P.; Mighton, J.; Lam, D.; Warren, D.; Stephenson, J.,
Cranberry juice consumption may reduce biofilms on uroepithelial cells: pilot study in
spinal cord injured patients. Spinal Cord 2001, 39, (1), 26-30.
116.
Reid, G., The role of cranberry and probiotics in intestinal and urogenital tract
health. Crit Rev Food Sci 2002, 42, (3), 293-300.
117
117.
Russell, W. R.; Labat, A.; Scobbie, L.; Duncan, S. H., Availability of blueberry
phenolics for microbial metabolism in the colon and the potential inflammatory
implications. Molecular Nutrition & Food Research 2007, 51, (6), 726-731.
118.
Heinonen, M., Antioxidant activity and antimicrobial effect of berry phenolics -
a Finnish perspective. Molecular Nutrition & Food Research 2007, 51, (6), 684-691.
119.
Villarini, M.; Caldini, G.; Moretti, M.; Trotta, F.; Pasquini, R.; Cenci, G.,
Modulatory activity of a lactobacillus casei strain on 1.2-dimethylhydrazine-induced
genotoxicity in rats. Environmental and Molecular Mutagenesis 2008, 49, (3), 192-199.
120.
Kim, D. H.; Jung, E. A.; Sohng, I. S.; Han, J. A.; Kim, T. H.; Han, M. J.,
Intestinal bacterial metabolism of flavonoids and its relation to some biological
activities. Archives of Pharmacal Research 1998, 21, (1), 17-23.
121.
Clifford, M. N.; Scalbert, A., Ellagitannins - nature, occurrence and dietary
burden. J Sci Food Agr 2000, 80, (7), 1118-1125.
122.
Stoner, G. D.; Wang, L. S.; Zikri, N.; Chen, T.; Hecht, S. S.; Huang, C.; Sardo,
C.; Lechner, J. F., Cancer prevention with freeze-dried berries and berry components.
Semin Cancer Biol 2007, 17, (5), 403-410.
123.
Boateng, J.; Verghese, M.; Shackelford, L.; Walker, L. T.; Khatiwada, J.; Ogutu,
S.; Williams, D. S.; Jones, J.; Guyton, M.; Asiamah, D.; Henderson, F.; Grant, L.;
DeBruce, M.; Johnson, A.; Washington, S.; Chawan, C. B., Selected fruits reduce
azoxymethane (AOM)-induced aberrant crypt foci (ACF) in Fisher 344 male rats. Food
Chem Toxicol 2007, 45, (5), 725-732.
124.
Seeram, N. P., Berries. Chapter 37. Nutritional
Oncology, 2nd ed.; Heber, D., Blackburn, G., Go, V. L.
W., Milner, J., Eds.; Academic Press: London, UK, 2006, 615-625.
125.
Seeram, N. P.; Adams, L. S.; Zhang, Y. J.; Lee, R.; Sand, D.; Scheuller, H. S.;
Heber, D., Blackberry, black raspberry, blueberry, cranberry, red raspberry, and
strawberry extracts inhibit growth and stimulate apoptosis of human cancer cells in
vitro. J Agr Food Chem 2006, 54, (25), 9329-9339.
126.
McDougall, G. J.; Ross, H. A.; Ikeji, M.; Stewart, D., Berry extracts exert
different antiproliferative effects against cervical and colon cancer cells grown in vitro.
J Agr Food Chem 2008, 56, (9), 3016-3023.
118
127.
Ross, H. A.; McDougall, G. J.; Stewart, D., Antiproliferative activity is
predominantly associated with ellagitannins in raspberry extracts. Phytochemistry 2007,
68, (2), 218-228.
128.
Mertens-Talcott, S. U.; Lee, J. H.; Percival, S. S.; Talcott, S. T., Induction of cell
death in Caco-2 human colon carcinoma cells by ellagic acid rich fractions from
muscadine grapes (Vitis rotundifolia). J Agr Food Chem 2006, 54, (15), 5336-5343.
129.
Yi, W. G.; Fischer, J.; Akoh, C. C., Study of anticancer activities of muscadine
grape phenolics in vitro. J Agr Food Chem 2005, 53, (22), 8804-8812.
130.
Lizarraga, D.; Lozano, C.; Briede, J. J.; van Delft, J. H.; Tourino, S.; Centelles,
J. J.; Torres, J. L.; Cascante, M., The importance of polymerization and galloylation for
the antiproliferative properties of procyanidin-rich natural extracts. Febs J 2007, 274,
(18), 4802-11.
131.
Lizarraga, D.; Tourino, S.; Reyes-Zurita, F. J.; de Kok, T. M.; van Delft, J. H.;
Maas, L. M.; Briede, J. J.; Centelles, J. J.; Torres, J. L.; Cascante, M., Witch Hazel
(Hamamelis virginiana) Fractions and the Importance of Gallate Moieties-Electron
Transfer Capacities in Their Antitumoral Properties. J Agr Food Chem 2008, 56, (24),
11675-11682.
132.
Tanimura, S.; Kadomoto, R.; Tanaka, T.; Zhang, Y. J.; Kouno, I.; Kohno, M.,
Suppression of tumor cell invasiveness by hydrolyzable tannins (plant polyphenols) via
the inhibition of matrix metalloproteinase-2/-9 activity. Biochem Bioph Res Co 2005,
330, (4), 1306-1313.
119
SUMMARY AND GENERAL CONCLUSIONS
Polyphenols identified in the edible part of mango from different varieties
(Haden, Ataulfo, Kent, Tommy Atkins and Francis) comprise gallic acid, mangiferin,
phenolic acid derivatives and a wide range of high molecular weight hydrolysable
tannins. Ataulfo and Haden polyphenols inhibited the growth of human SW-480 colon
cancer cells, which showed high sensitivity to the treatment. Additionally, non-cancer
CCD-18Co colon cells showed lower sensitivity to polyphenols treatment than human
cancer colon cells. SW-480 gene regulation influenced by Ataulfo and Haden
polyphenols suggested the induction of apoptosis through intrinsic and extrinsic
mechanisms, in addition to cell cycle arrest. Cell cycle arrest in the G2/M phase was
related to the upregulation of PKMYT1 gene expression. Ataulfo and Haden
polyphenols exerted protection of non-cancer CCD-18Co colon cells by lowering the
ROS generation in a dose dependent manner. Based on their anti-cancer effects and
protective effects in normal cells, but not in cancer cells, mango polyphenolic extracts
may have a high potential as chemopreventive agent.
The absorption of mango polyphenols was predominantly characterized by low
molecular weight compounds, mainly phenolic acids in their free forms like gallic,
caffeic, ferulic, p-OH- coumaric and vanilic acids, with no evidence of absorption of
high molecular weight polyphenols like mangiferin (422 Da) and gallotannins with
molecular weight ranging from 788 Da (tetra-O-galloyl-glucose) to 1852 Da (undeca-Ogalloyl-glucose). Despite enhanced absorption facilitated by enzymatic hydrolysis, a
significant increase in antioxidant activity, phenolic content and antiproliferative effects
on MDA-MB-231 breast and HT-29 colon cancer cells was not observed. Additionally,
both high (422; 788-1852 Da) and low (138-194 Da) molecular weight polyphenols
rich-fractions equally inhibited cell proliferation of colon and breast cancer cells at the
same extent (0-20 μg of gallic acid equiv/mL), which may indicate that the anti-cancer
efficacy of mango polyphenolics is not dependent on enzymatic hydrolysis.
These results corroborate previous findings from in vivo studies, which suggest
that the most of mango polyphenols are not absorbed intact through the small intestine,
but may be hydrolyzed by intestinal enzymes into low molecular weight aromatic acids,
which would be later absorbed; or when polyphenols are not absorbed, they likely reach
120
the large intestine, modulating the gut microflora, and thus they contribute to reduce the
risk of colon carcinogenesis.
Overall, polyphenols from several mango varieties exerted anti-cancer effects,
and these effects may not require enzymatic hydrolysis by β-glucosidase.
Although anti-cancer properties of mango pulp polyphenols was demonstrated,
additional in vivo and in vitro studies regarding the chemopreventive mechanisms are
necessary to better elucidate the anti-cancer properties of mango polyphenols.
121
APENDIX
Table 1. HPLC-ESI-MSn of mango pulp polyphenols extracted from the varieties
Ataulfo (A), Kent (B), Tommy Atkins (C), Francis (D) and Haden (E).
(A)
RT: 14.04 - 44.56
NL:
1.85E5
Total Scan
PDA
EnzymeTes
t_Stock
37.57
100
38.06
17.71
90
80
36.80
17.04
70
36.42
Relative Absorbance
60
50
36.01
25.81
40
26.95
30
22.92
35.32
28.44
29.09
20
42.17
33.95
10
32.88
19.43
0
-10
-20
15
Peak
20
Identity
25
RT(min)
30
Time (min)
HPLC-DAD
λmax [nm]
1
mono-O-galloylglucose
17.71
237,278
2
mangiferin
28.46
209,237,259,318,365
3
penta-O-galloylglucose
33.95
238,277
4
hexa-O-galloylglucose
35.32
238,277
5
hepta-O-galloylglucose
35.79
238,276
122
35
40
HPLC-ESI(-)-MS2
(m/z) (% base peak)
MS2 [331.14]: 271.03 (100),
169.10(86.48), 223.05
(40.84)
MS2 [421.02]: 301.21 (100),
331.13(95.64), 403.06
(25.63)
MS2 [938.88]: 768.92 (100),
786.85 (21.23)
MS2 [1090.66]:
1090.97(100),
1191.92(97),958.61(84.31),
1198.45(69.63),938.93(54)
MS2 [1242.64]:
1016.47(100),
996.22(63.47),902.56(61.88),
1220.62(57.16)
6
octa-O-galloyl- glucose
36.08
238,276
7
nona-O-galloylglucose
37.51
239,274
8
deca-O-galloylglucose
38.17
239,274
9
undeca-O-galloylglucose
39.21
239,274
MS2 [1394.59]:
1090.73(100),
1242.54(77.07),
938.76(10.23)
MS2 [1546.56]:
1242.61(100),
1394.55(61.33),
1090.74(26.67)
MS2 [1698.46]:
1394.53(100),
1242.64(60.31),
1546.48(48.92)
1090.68(17.14)
MS2 [1850.32]:
1546.49(100),
1394.56(84.88)
(B)
RT: 0.00 - 60.00
19.17
100
NL:
5.43E5
Total Scan
PDA
Michelle_K
ent1
25.68
90
80
70
Relative Absorbance
22.73
60
32.57
36.27
45.58
26.33
50
32.32
40
37.42
29.40
30
37.95
38.97
18.40
20
40.50
17.39
10
5.11 8.76 10.72
0
0
5
10
41.38
45.99
57.26
59.40
15.54
15
20
25
30
Time (min)
123
35
40
45
50
55
60
Peak
Identity
RT(min)
HPLC-DAD
λmax [nm]
HPLC-ESI(-)-MS2
(m/z) (% base peak)
MS2 [331.19]: 271.05
(100),169 (84.3)
1
mono-O-galloylglucose
19.25
232,271
2
tetra-O-galloylglucose
31.40
232,275
MS2 [787.02]: 617.04 (100),
635.07 (42.04)
3
penta-O-galloylglucose
33.39
232,274
MS2 [939.05]: 769.04 (100),
787.08 (23.37)
4
hexa-O-galloylglucose
34.72
232,277
MS2 [1090.90]: 919.07(100),
920.28(52.32),770.34(33.90)
5
hepta-O-galloylglucose
35.69
232,277
6
octa-O-galloyl- glucose
36.43
232,275
7
nona-O-galloylglucose
37.92
232,272
MS2 [1242.47]:
1090.17(100),
938.21(47.39),1090(24.39),
769.70(17.34)
MS2 [1394.80]:
1241.41(100), 939.91(65.05),
938.03(33.67)
MS2 [1547.67]:
1242.78(100),
1090.86(65.14)
1394.76(39.79)
(C)
RT: 15.97 - 46.45
NL:
4.87E5
Total Scan
PDA
Michelle_To
mmy1_090
506150025
26.86
100
90
19.12
22.72
80
Relative Absorbance
70
26.35
25.68
60
36.29
50
40
36.00
31.11
20.77
30
25.31
35.11
20
10
29.42
32.60
45.59
38.58
38.95
18.07
40.47
24.17
17.78
37.14
40.99
0
20
25
30
Time (min)
124
35
40
45
Peak
Identity
RT(min)
HPLC-DAD
λmax [nm]
HPLC-ESI(-)-MS2
(m/z) (% base peak)
MS2 [331.19]: 169 (100)
1
mono-O-galloylglucose
19.25
232,279
2
penta-O-galloylglucose
33.0
232,278
MS2 [939.11]: 769.08 (100),
787.05 (21.30)
3
hexa-O-galloylglucose
34.11
232,277
MS2 [1090.78]: 928.08 (100),
769.92(53.95)
4
hepta-O-galloylglucose
35.11
232,274
5
octa-O-galloylglucose
36.29
232,275
6
nona-O-galloylglucose
37.42
232,279
MS2 [1242.81]:
938.13(100),1014.16(88.90),
1088.93(26.34)
MS2 [1394.77]: 1241.89(100),
1089.90(51.81),
1241.10(39.06),1089.13(38.13),
939.96(30.40),
MS2 [1546.75]: 1240.77(100),
1392.84(64.45),
1393.70(53.19),
1241.79(92.42), 1242.72(39.50)
(D)
RT: 0.00 - 60.00
NL:
5.40E5
Total Scan
PDA
Michelle_Fr
ancine1
19.14
100
90
80
26.95
RelativeAbsorbance
70
60
36.30
24.27
37.49
50
31.14
40
18.34
45.63
35.05
30
18.01
38.59
39.01
20
17.70
40.54
41.42
17.39
10
3.82
0
0
8.78
5
10.75
10
48.08
57.33
59.39
15.27
15
20
25
30
Time (min)
125
35
40
45
50
55
60
Peak
Identity
RT(min)
HPLC-DAD
λmax [nm]
HPLC-ESI(-)-MS2
(m/z) (% base peak)
MS2 [331.14]: 169 (100)
1
mono-O-galloylglucose
19.25
232,279
2
tetra-O-galloylglucose
29.44
232,272
MS2 [787.03]: 617.04 (100),
635.03 (17.23)
3
penta-O-galloylglucose
32.92
232,278
MS2 [939.05]: 769.06 (100),
787.00 (24.79)
4
hexa-O-galloylglucose
34.17
232,278
MS2 [1090.88]: 938.04 (100),
769.66(93.03),
768.23(50.27),752.40(47.72)
5
hepta-O-galloylglucose
35.17
232,277
MS2 [1242.81]: 1088.99(100)
949.98(74.94),1027.99(43.27),
6
octa-O-galloylglucose
36.08
232,274
MS2 [1394.88]: 1241.59(100),
1089.89(37.40),
1373.08(24.30), 937.99(17.01)
7
nona-O-galloylglucose
37.19
232,275
MS2 [1547.76]: 1242.78(100),
1394.78(41.77), 1090.79(55.46)
(E)
RT: 0.00 - 60.00
NL:
4.82E5
Total Scan
PDA
Michelle_H
aden1_090
506121412
26.90
100
95
90
19.06
85
80
20.74
75
70
Relative Absorbance
65
60
55
26.38
50
29.44
36.36
25.72
45
45.70
40
35
30
25.36
18.30
37.20
25
17.98
20
24.19
39.07
17.56
15
10
40.62
41.03
15.52
46.14
54.81
56.77
9.81
5
0.89
0
0
5.38
5
13.00
10
15
20
25
30
Time (min)
126
35
40
45
50
55
60
Peak
Identity
RT(min)
HPLC-DAD
λmax [nm]
HPLC-ESI(-)-MS2
(m/z) (% base peak)
MS2 [331.91]: 169 (100)
1
mono-O-galloylglucose
19.06
232,279
2
di-O-galloyl- glucose
25.58
232,281
MS2 [634.96]: 483.07 (100),
465.08 (21.26)
3
tetra-O-galloylglucose
29.23
232,272
MS2 [787.10]: 617.05 (100),
635.07 (51.85)
4
mangiferin
30.31
232,258,361
MS2 [421.18]: 301.24 (100),
331.14 (91.64)
5
penta-O-galloylglucose
33.01
232,278
MS2 [939.05]: 769.04 (100),
787.06 (14.73)
6
hexa-O-galloylglucose
35.16
232,278
MS2 [1090.81]: 928.16 (100),
769.06(79.53),
806.46(62.11),769.67(34.24)
7
hepta-O-galloylglucose
35.32
232,277
MS2 [1242.81]: 1088.99(100)
949.98(74.94),1027.99(43.27),
232,274
MS2 [1395.14]: 1242.86(100),
1241.87(24.26),
1090.83(77.86),
1373.08(24.30), 938.95(44.95)
8
octa-O-galloylglucose
36.20
127
Download

MICHELE CORRÊA BERTOLDI ANTIOXIDANT CAPACITY